WO2016103531A1 - Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug - Google Patents
Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug Download PDFInfo
- Publication number
- WO2016103531A1 WO2016103531A1 PCT/JP2014/084772 JP2014084772W WO2016103531A1 WO 2016103531 A1 WO2016103531 A1 WO 2016103531A1 JP 2014084772 W JP2014084772 W JP 2014084772W WO 2016103531 A1 WO2016103531 A1 WO 2016103531A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- spg
- seq
- cells
- oligodeoxynucleotide
- tumor
- Prior art date
Links
Images
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/7125—Nucleic acids or oligonucleotides having modified internucleoside linkage, i.e. other than 3'-5' phosphodiesters
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/715—Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
- A61K31/716—Glucans
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
- A61K47/549—Sugars, nucleosides, nucleotides or nucleic acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/56—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
- A61K47/61—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule the organic macromolecular compound being a polysaccharide or a derivative thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Definitions
- the present invention relates to a novel cancer treatment.
- CpG ODN CpG oligonucleotide
- TLR9 Toll-like receptor 9
- DCs dendritic cells
- IFNs type I interferons
- CTL cytotoxic T lymphocyte
- Non-patent Document 6 There are at least four types of CpG ODN, each having a different skeletal sequence and immunostimulatory properties (Non-patent Document 6).
- D-type (also referred to as A-type) CpG ODNs typically contain one palindromic CpG motif with a phosphodiester (PO) backbone and a phosphorothioate (PS) poly-G tail, and plasmacytoid DCs (pDCs) Is activated to produce a large amount of IFN- ⁇ , but cannot induce pDC maturation or B cell activation (Non-patent Documents 7 and 8).
- the other three types of ODN consist of a PS skeleton.
- K-type (also referred to as B-type) CpG ODN typically contains multiple CpG motifs of non-palindrome structure and strongly activates B cells to produce IL-6 and activates pDCs
- IFN- ⁇ is hardly produced (Non-patent Documents 8 and 9).
- Recently developed C-type and P-type CpG ODNs contain one and two palindromic CpG sequences, both of which activate B cells like K type and like D type
- pDCs can be activated
- C-type CpG ODN induces IFN- ⁇ production weaker than P-type CpG ODN (Non-patent Documents 10-12).
- Patent Document 1 describes many excellent K-type CpG ODNs.
- D-type and P-type CpG ODNs are Hoogsteen base pairs that form parallel four-stranded structures called G-tetrads, and Watson-Crick base pairs between cis and trans palindrome structures, Respectively, which are necessary for strong IFN- ⁇ production by pDCs (Non-patent Documents 12-14).
- Such higher-order structures appear to be necessary for early endosome localization and TLR9-mediated signaling, but these are affected by product polymorphism and precipitation, thus preventing its clinical application (Non-patent document 15). Therefore, only K-type and C-type CpG ODN are generally available as human immunotherapeutic agents and vaccine adjuvants (Non-patent Documents 16 and 17).
- K-type CpG ODN enhances the immunogenicity of vaccines targeting infectious diseases and cancer in human clinical trials (Non-Patent Documents 6 and 16), but for optimal adjuvant effect, antigen and K-type CpG Chemical and physical linkage between the ODN is required.
- Schizophyllan a soluble ⁇ -1,3-glucan derived from Schizophyllum commune, is a drug approved in Japan for 30 years as an activator of radiation therapy in cervical cancer patients.
- Patent Document 18 Lentinan (LNT), a soluble ⁇ -1,3-glucan derived from shiitake mushroom, was approved in 1985 and used in combination with fluoropyrimidines for patients with inoperable and recurrent gastric cancer. (Non-Patent Documents 19 and 20). It has been shown that ⁇ -1,3-glucan forms a complex of triple helix structure with polydeoxyadenylic acid (dA) (Non-patent Document 21).
- dA polydeoxyadenylic acid
- Patent Documents 2 to 4 disclose the use of a water-soluble complex of ⁇ -1,3-glucan containing schizophyllan and a nucleic acid (gene) as a gene carrier. These documents describe that the antisense action of a gene and the resistance action to a nucleolytic enzyme (nuclease) are enhanced by forming the complex.
- Patent Document 5 by using a polysaccharide having ⁇ -1,3-linkage as a carrier (transfection agent), a CpG sequence is contained, and a phosphodiester bond is substituted with a phosphorothioate bond or a phosphorodithioate bond. It is disclosed that the action of immunostimulatory oligonucleotides is enhanced.
- Patent Document 6 describes an immunostimulatory complex comprising an immunostimulatory oligonucleotide and ⁇ -1,3-glucan having a long ⁇ -1,6-glucoside-binding side chain. Has been.
- Non-patent Documents 22 and 23, Patent Document 7 disclose the present inventors that mouse and humanized CpG ODN, which were conjugated with poly (dA) having a phosphodiester bond at the 5 ′ end, complexed with SPG, enhanced cytokine production and influenza vaccine. It has been shown to act as an adjuvant or a preventive or therapeutic agent for Th2 cell-related diseases.
- poly (dA) was added to the 5 'end of each of the K-type and D-type CpG to form a complex with SPG, the activity was enhanced while maintaining the properties of the K-type and D-type, respectively.
- Non-patent Document 24 In recent years, it has been shown that when poly (dA) having a phosphorothioate bond is linked to CpG ODN, complex formation is increased to almost 100% (Non-patent Document 24). However, no thorough testing has been done to identify the optimal humanized CpG sequence and optimize the factors to obtain the “all-in-one” activity of the four types of CpG ODN.
- Patent Document 8 discloses a method for producing an antigen / CpG oligonucleotide / ⁇ -1,3-glucan ternary complex.
- CpG ODN Synthetic nucleic acid CpG oligodeoxynucleotide
- TLR9 Toll-like receptor 9
- CpG ODN is also expected as an immunotherapeutic agent for cancer.
- conventional CpG ODN has antitumor activity, it can only exert its effect by being administered directly to the tumor, and it has been thought that clinical application is difficult. In fact, it may be difficult to administer a drug directly to a tumor at an early stage in clinical practice. In addition, surgical treatment is required in the deep part, and the hurdle is high.
- K3-SPG TLR9 ligand
- PCT application PCT / JP2014 / 0748835
- K3-SPG has been shown by experiments using mice to activate innate immunity more strongly than conventional CpG ODN without forming aggregates, and at the same time a strong adjuvant effect.
- K3-SPG induces strong acquired immunity not only in mice but also in cynomolgus monkeys, and it was possible to overcome the difference in reactivity between mice and primates, which had been feared so far.
- Non-Patent Document 29 Kang, X., et al.The Journal of Immunology 155,1343-1348 (1995)).
- Cancer immunotherapy which has received particular attention, is a cancer vaccine that uses antigen-presenting cells of autologous peripheral blood that was first approved by the US Food and Drug Administration (FDA) in April 2010 for prostate cancer patients.
- Non-patent document 30 Cancer vaccine approval couled open floodgates.Nature medicine 16,615-615 (2010);
- Non-patent document 31 Higano, CS, et al. Cancer 115, 367079) ).
- Non-Patent Document 32 Phan, GQ, et al. Proc. Natl. Acad. Sci. USA 100, 8372-8377 (2003);
- Non-Patent Document 33 Camacho, L. et al. H., et al. Journal of clinical oncology: official journal of the American Society of Clinical Oncology 27, 1075-1081 (2009):
- Non-patent document 34 Hodi. land Journal of Medicine 363,711-723 (2010)).
- Non-patent Document 35 AZIJLI, K., et al. Anticancer Research 34, 1493- 1505 (2014);
- Non-Patent Document 36 Okazaki, T., et al. Nature immunology 14, 1212-1218 (2013);
- Non-Patent Document 37 Ishida, Y., et al. The EMBO journal 11, 3877-3895. (1992);
- Non-Patent Document 38 Topalian, SL, et al. The New England journal of medicine 366, 2443-2454 (2012)).
- Non-Patent Document 43 Garaude, J., et al. Science translational medicine 4,120ra116 (2012);
- Non-Patent Document 44 Martinez-Pomares. , L. et al. Trends in immunology 33, 66-70 (2012)).
- Non-Patent Document 45 Palucka, K. et al. Nature reviews. Cancer 12, 265-277 (2012)).
- K3-SPG a complex of human K-type CpG ODN K3 and beta glucan
- K3-SPG confirmed tumor regression in tumor-bearing mice after intravenous administration, which was not effective with conventional CpG ODN (K3) (FIG. 2 (A ⁇ )).
- K3-SPG confirmed tumor regression in tumor-bearing mice after intravenous administration, which was not effective with conventional CpG ODN (K3) (FIG. 2 (A ⁇ )).
- the present invention has been completed, and the present inventors have further demonstrated that a peritoneal dissemination model, which is a more clinical model, also exhibits strong antitumor activity (FIG. 2g, m (FIG. 2)). 2B))
- the present inventors did not require antigen administration for this effect, and confirmed the effect by administration of a single agent.
- the present inventors have found that the acquired immune response is important for the antitumor effect of K3-SPG and that the type I interferon (IFN) and IL-12 induced by the innate immune response are essential. Shown using mice (FIGS. 6a, b, c (FIG. 6A)). In addition, the present inventors confirmed that CD45 negative tumor cells were accumulated in the spleen by intravenous administration of K3-SPG, and many of these cells caused cell death (necrosis or apoptosis). I made it clear.
- mice When mice were immunized with CD45-negative cells, they exerted a strong antitumor effect, and thus it was clarified that cell death of CD45-negative cells accumulated in the spleen appears to play an important role ( 6g, h, i, j (FIG. 6B)).
- CD8T cells activated in tumors accumulate by administering K3-SPG, and it is clear that these cells are essential for antitumor effects. It was.
- CpG ODN that exerts an antitumor effect in systemic administration is expected to work strongly even in carcinomas that have been difficult so far. Furthermore, since CpG ODN exhibits an antitumor effect without an antigen, application as a single agent can also be expected.
- CpG ODN has been treated as a single agent (Platesi, G., et al. Cancer research 65, 6388-6393 (2005); Manegold, C., et al. Annals ofology. Medical Oncology / ESMO 23, 72-77 (2012); Kim, YH, et al. Blood 119, 355-363 (2012); Hirsh, V., et al. Journal of clinical onology: American Society of Clini al Oncology 29, 2667-2675 (2011); Weber, JS, et al. Cancer 115, 3944-3594 (2009) or cancer vaccine adjuvant (Reed, SG, Nature medicine 19, 1597-). 1608 (2013); Perret, R., et al.
- Clinical cancer research an official journalAmericanCertificateAmerica , 6516-6522 (2003); Lou, Y., et al., Journal of immu. other (Hagerstown, Md .: 1997) 34, 279-288 (2011); Auf, G., Clinical cancer research: an official journal of the American Assoc. , Et al. PLoS One 4, e8368 (2009); Heckelsmiller, K., et al. Journal of immunology 169, 3892-3899 (2002)).
- K3-SPG nanoparticulate TLR9 agonist
- SPG schizophyllan
- K3-G B / K type CpG
- the present inventors further examined the potential of K3-SPG single-agent immunotherapy for cancer (without using additional tumor peptides and antigens) and found that the above effects were obtained. As a result, the present invention was completed. Therefore, the present invention typically provides the following.
- Anticancer agent alone (1) (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is arranged on the 3 ′ side of the humanized K-type CpG oligodeoxynucleotide An oligodeoxynucleotide, (B) An anticancer agent containing a complex containing ⁇ -1,3-glucan. (2) The anticancer agent according to item (1), wherein the anticancer agent is administered without a cancer antigen.
- the reticuloendothelial system and / or lymph node includes a tumor and a phagocytic cell.
- the systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
- the oligodeoxynucleotides are K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5)
- the anticancer agent according to any one of items 1 to 8, selected from the group consisting of K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 12 ⁇ 7).
- the ⁇ -1,3-glucan is any one of items 1 to 9, wherein the ⁇ -1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran, and laminaran.
- Anticancer drugs are selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran, and laminaran.
- SPG schizophyllan
- lentinan lentinan
- scleroglucan scleroglucan
- curdlan e.g., curdlan
- parkan e.g., parkan
- glyphoran glyphoran
- laminaran glyphoran
- (Reticuloendothelial system (including spleen and / or liver) and / or lymph node accumulating agent) (12) (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide An oligodeoxynucleotide, (B) A composition for accumulating dead cells of cancer in the spleen, comprising a complex containing ⁇ -1,3-glucan.
- the oligodeoxynucleotide K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5)
- the composition according to item (12) selected from the group consisting of K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- the ⁇ -1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran, according to item (12) or (13) Composition.
- SPG schizophyllan
- lentinan lentinan
- scleroglucan curdlan
- parkan glyphoran
- laminaran laminaran
- composition according to any one of items (12) to (17), wherein the administration comprises systemic administration comprises systemic administration.
- systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
- the oligodeoxynucleotides are K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5)
- the composition according to item (20) which is K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- the ⁇ -1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran, according to item (20) or (21) Composition.
- SPG schizophyllan
- lentinan lentinan
- scleroglucan curdlan
- parkan glyphoran
- laminaran laminaran
- K3-SPG of the present invention as an antitumor agent can exert a strong antitumor effect in systemic administration that could not be overcome by conventional CpG ODN. Therefore, it is considered to be very useful from a clinical viewpoint.
- sufficient effects innate immune response
- Our research results show that it has a strong antitumor effect in addition to the very strong innate immune activation ability compared to CpG ODN used in clinical trials so far.
- SPG can be expected as a useful immunotherapy drug.
- the effect can be exerted by inducing cell death of tumor cells without requiring administration of an antigen, it can be applied to various carcinomas. From these results, K3-SPG has the potential as an innate immune activated antitumor drug that does not require an antigen.
- FIG. 1 shows a method of complexing SPG with CpG ODN.
- FIG. 2 (AB) shows that systemic injection of antigen-free nanoparticulate CpG (K3-SPG) can be applied to many established tumor models, including pancreatic cancer peritoneal dissemination models. Show.
- FIG. 2A shows ai. C57BL / 6 mice were treated with EG7 cells on day 0. c.
- FIG. 2 shows that systemic injection of antigen-free nanoparticulate CpG (K3-SPG) can be applied to many established tumor models, including pancreatic cancer peritoneal dissemination models. Show. FIG. 2B shows j to n.
- mice (J to l) C57BL / 6 mice were inoculated with B16 cells, B16F10 cells or MC38 cells on day 0.
- the B16 inoculated group was challenged i.e. on days 10, 12, and 14 with K3-SPG. v. Or i. t. Treated.
- the B16F10 inoculated group was challenged i.e. on days 7, 9 and 11 with K3-SPG. v. Or i. t. Treated.
- K3-SPG can be a cancer immunotherapy agent that does not require an antigen.
- EG7 which is a tumor cell line
- K3 and K3-SPG were intravenously administered three times (7th, 9th, and 11th days). Tumor size was measured from day 7 after transplantation of tumor cells.
- FIG. 4 shows that K3-SPG targets phagocytic cells within the tumor microenvironment.
- Ac C57BL / 6 mice were treated with EG7 on day 0.
- mice were analyzed with an in vivo fluorescence imaging system (IVIS) and the image measured with relative fluorescence was converted to physical units of surface radiance (photons / second / cm 2 / sr).
- IVIS in vivo fluorescence imaging system
- White arrows indicate the tumor inoculation area (a).
- FIG. 6A Cryosections of tumors from FIG. 6a (FIG. 6A) were stained with anti-CD3e antibody (red, EG7 staining) and Hoechst 33258 (blue, nuclear staining) and then analyzed with a fluorescence microscope (scale bar) , 100 ⁇ m).
- White arrows indicate fluorescence positive areas.
- FIG. 5 shows that F4 / 80 positive cells in the tumor were depleted by clodronate liposomes.
- C57BL / 6 mice were inoculated with EG7 on day 0 and clodronate liposomes (a) or control liposomes (b) were injected i. v. And on day 7 with Alexa 647-K3-SPG i. v. Treated.
- FIG. 6 (AB) shows that both IL-12 and IFN are important for their potential role in tumor shrinkage and their immunogenic cell death.
- FIG. 6A shows a to f.
- K Tumor volume at day 25 and the number of OVA 257-264 specific tetramers + CD8 T cells are represented by bar graphs and scatter paper diagrams, respectively. * P ⁇ 0.05 (t test).
- FIG. 7 shows that IFN- ⁇ was detected in the tumor microenvironment.
- IFN- ⁇ GFP mice were inoculated with EG7 on day 0 and on days 7, 9, and 11 with K3-SPG. d. Or i. v. Treated. Twelve days after inoculation, tumors were collected and frozen sections were stained with anti-CD11b antibody, anti-CD169 antibody, anti-F4 / 80 antibody, anti-MARCO antibody (red) and Hoechst 33258 (blue) and then analyzed with a fluorescence microscope. (Scale bar, 100 ⁇ m).
- IFN- ⁇ positive cells were counted (10 fields from each of 3 tumors). Error bars represent mean + SD. * P ⁇ 0.05 (t test).
- FIG. 8 shows that IL12-p40 was detected in the tumor microenvironment.
- A C57BL / 6 mice were inoculated with EG7 on day 0 and on days 7, 9, and 11 with K3-SPG. d. Or i. v. Treated. Twelve days after inoculation, tumors were collected and frozen sections were stained with anti-IL12-p40 antibody (red) and Hoechst 33258 (blue) and then analyzed with a fluorescence microscope (scale bar, 100 ⁇ m).
- B IL12-p40 positive cells were counted (10 fields from each of 3 tumors). Error bars represent mean + SD. * P ⁇ 0.05 (t test).
- FIG. 9 shows that CD45 negative cells are derived from tumor cells but not from host cells.
- FIG. 10 shows that K3-SPG-induced tumor shrinkage is innate and adaptive immunity including Il12, type 1 IFN, Batf3, CD8 + DC, and potent cytotoxic T cells infiltrating the tumor. Indicates that both responses are required.
- FIG. 10A shows a to c.
- C57BL / 6 knockout mice (a), as well as Batf3 heterozygous and Batf3 knockout mice (b), were inoculated with EG7 cells on day 0 and i.v. K3-SPG on days 7, 9, and 11.
- v. Treated black arrow.
- C C57BL / 6 mice were inoculated with EG7 on day 0 and on days 7, 9 and 11 with K3-SPG i. d. Or i. v. Treated with.
- FIG. 10 shows that K3-SPG-induced tumor shrinkage is innate and adaptive immunity including Il12, type 1 IFN, Batf3, CD8 + DC, and potent cytotoxic T cells infiltrating the tumor. Indicates that both responses are required.
- FIG. 10B shows de.
- FIG. 11 shows a schematic paper diagram of the experimental system.
- WT and Il12p40-Ifnar2 DKO mice are inoculated with EG7 cells on day 0 and on days 7, 9 and 11 with K3-SPG or PBS.
- v. Treated. On day 14, another CD8 ⁇ + T cell was purified from the spleen of these mice, labeled with Xenlight DiR®, and treated with K3-SPG (days 7, 9 and 11).
- FIG. 12 shows the K3-SPG treatment strategy.
- K3-SPG targeted the tumor microenvironment via the bloodstream.
- K3-SPG targeted phagocytic cells and activated these cells.
- IFN and IL-12 were induced by K3-SPG treatment.
- the antigen was then released via lymph flow and blood flow. The presentation of this antigen induced a strong tumor-specific CTL.
- the present invention provides oligodeoxynucleotides (hereinafter referred to as oligodeoxynucleotides of the present invention) containing K-type CpG oligodeoxynucleotides and polydeoxyadenylic acid (dA).
- oligodeoxynucleotides of the present invention include those in which phosphodiester bonds are modified (for example, some or all of the phosphodiester bonds are replaced by phosphorothioate bonds).
- the oligodeoxynucleotides of the present invention include pharmaceutically acceptable salts.
- CpG oligonucleotide (residue) or “CpG oligodeoxynucleotide (residue)”, “CpG ODN (residue)” or simply “CpG (residue)” are used interchangeably.
- a polynucleotide preferably an oligonucleotide, comprising at least one unmethylated CG dinucleotide sequence, which is synonymous with or without the term “residue” at the end.
- An oligonucleotide comprising at least one CpG motif can comprise multiple CpG motifs.
- CpG motif refers to an unmethylated dinucleotide portion of an oligonucleotide comprising a cytosine nucleotide followed by a guanosine nucleotide. 5-methylcytosine can also be used in place of cytosine.
- polydeoxyadenylic acid and polydeoxyadenosine acid are synonymous.
- the term “residue” means a partial structure of a compound with a higher molecular weight, but in this specification “CpG oligodeoxynucleotide (CpG ODN)” means an independent molecule or a compound with a higher molecular weight. The meaning of the partial structure is easily understood by those skilled in the art from the context. The same applies to terms relating to other partial structures contained in the oligodeoxynucleotide of the present invention such as “polydeoxyadenylic acid”.
- CpG ODN CpG oligonucleotide
- TLR9 Toll-like receptor 9
- DCs dendritic cells
- B cells B cells to produce type I interferons (IFNs) and inflammatory cytokines (Hemmi, H., et al. Nature 408, 740-745 (2000); Krieg , AM Nature reviews.
- Drug discovery 5,471-484 (2006) acts as an adjuvant for Th1-type humoral and cellular immune responses, including cytotoxic T lymphocyte (CTL) reactions ( Brazolot Millan, CL, Weera na, R., Krieg, AM, Siegrist, CA & Davis, HL Proceedings of the National Academy of Sciences of the United States3, 15A15, 15:15 (America). RS, Targoni, OS, Krieg, AM, Lehmann, PV & Harding, CV The Journal of experimental medicine 186, 1623-1631 (1997). Therefore, CpG ODN has been regarded as a potential immunotherapeutic agent for infectious diseases, cancer, asthma and hay fever (Krieg, AM Nature reviews. Drug discovery 5,471-484 (2006). Klinman, DM Nature reviews. Immunology 4, 249-258 (2004)).
- CTL cytotoxic T lymphocyte
- CpG ODN CpG oligodeoxynucleotide
- K type also called B type
- D type also called A type
- C type and P type Advanced drug delivery reviews 61
- K-type CpG ODNs contain multiple unmethylated CpG motifs, typically non-palindromic, and activate B cells to produce IL-6, but plasmacytoid dendritic cells (pDCs) It is a CpG ODN having structural and functional properties that hardly induce IFN- ⁇ production.
- An unmethylated CpG motif refers to a short nucleotide sequence containing at least one cytosine (C) -guanine (G) sequence, wherein the cytosine 5-position in the cytosine-guanine sequence is not methylated.
- CpG means unmethylated CpG unless otherwise specified.
- the oligodeoxynucleotide of the present invention contains K-type CpG ODN, thereby activating immunostimulatory activity peculiar to K-type CpG ODN (for example, B cells (preferably human B cells) and IL-6 Activity).
- K-type CpG ODN for example, B cells (preferably human B cells) and IL-6 Activity.
- a number of humanized K-type CpG ODNs are known in the art (Journal of immunology 166, 2372-2377 (2001); Journal of immunology 164, 944-953 (2000); US 8,030,285B2).
- the K-type CpG ODN contained in the oligodeoxynucleotide of the present invention is preferably humanized. “Humanized” means having agonist activity against human TLR9.
- the oligodeoxynucleotide of the present invention containing humanized K-type CpG ODN has an immunostimulatory activity peculiar to K-type CpG ODN (for example, an activity to activate human B cells to produce IL-6).
- K-type CpG ODN suitably used in the present invention has a length of 10 nucleotides or more and has the formula:
- N1, N2, N3, N4, N5 and N6 may be any nucleotide
- the K-type CpG ODN of the present invention is 10 nucleotides or more in length and includes a nucleotide sequence of the above formula.
- the central 4-base CpG motif (TCpGW) only needs to be contained in 10 nucleotides, and does not necessarily need to be located between N3 and N4 in the above formula.
- N1, N2, N3, N4, N5 and N6 may be any nucleotide, and N1 and N2, N2 and N3, N3 and N4, N4 and N5, and at least one of N5 and N6
- One (preferably one) combination may be a two-base CpG motif.
- any two consecutive bases in the central 4 bases (4th to 7th bases) are CpG motifs,
- the two bases can be any nucleotide.
- the K-type CpG ODN more preferably used in the present invention contains a non-palindrome structure containing one or more CpG motifs. Further preferably used K-type CpG ODN has a non-palindrome structure containing one or more CpG motifs.
- Humanized K-type CpG ODN is generally characterized by a 4-base CpG motif consisting of TCGA or TCGT. In many cases, two or three of these 4-base CpG motifs are contained in one humanized K-type CpG ODN. Therefore, in a preferred embodiment, the K-type CpG ODN contained in the oligodeoxynucleotide of the present invention has at least one, more preferably two or more, more preferably two or three, a 4-base CpG motif consisting of TCGA or TCGT. including. When the K-type CpG ODN has 2 or 3 4-base CpG motifs, these 4-base CpG motifs may be the same or different. However, there is no particular limitation as long as it has agonist activity against human TLR9.
- the K-type CpG ODN contained in the oligodeoxynucleotide of the present invention more preferably comprises the nucleotide sequence represented by SEQ ID NO: 1.
- the length of K-type CpG ODN is particularly limited as long as the oligodeoxynucleotide of the present invention has immunostimulatory activity (for example, the activity of activating B cells (preferably human B cells) to produce IL-6). Although not preferred, it is preferably no more than 100 nucleotides long (eg, 10-75 nucleotides long).
- the length of K-type CpG ODN is more preferably 50 nucleotides or less (for example, 10-40 nucleotides).
- the length of K-type CpG ODN is more preferably 30 nucleotides or less (for example, 10-25 nucleotides).
- the length of K-type CpG ODN is most preferably 12-25 nucleotides long.
- the length of polydeoxyadenylic acid (dA) is particularly limited as long as it is long enough to form a triple helical structure with a ⁇ -1,3-glucan (preferably lentinan or schizophyllan) chain.
- a ⁇ -1,3-glucan preferably lentinan or schizophyllan
- Poly dA forms a stable triple helix structure with ⁇ -1,3-glucan the longer it is, so there is theoretically no upper limit, but if it is too long, the length at the time of synthesis of oligodeoxynucleotides will vary.
- the length of poly dA is preferably 20 to 60 nucleotides (specifically, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 nucleotides long), more preferably 30 to 50 nucleotides long (30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41) , 42 , 43, 44, 45,
- the oligodeoxynucleotide of the present invention has an activity of forming a triple helical structure with two schizophyllan chains by including poly dA.
- polydeoxyadenylic acid may be described as “poly (dA)” or “poly (dA)”.
- One molecule of the oligodeoxynucleotide of the present invention may contain a plurality of K-type CpG ODN and / or poly dA, but preferably contains one K-type CpG ODN and poly dA one by one, Most preferably, each of K-type CpG ODN and poly dA consists of one by one.
- the oligodeoxynucleotide of the present invention is characterized in that poly dA is arranged on the 3 'side of K-type CpG ODN. With this arrangement, it is considered that the complex of the present invention (details will be described below) may also enhance the anticancer activity, but the present invention is not limited thereto. You may let them.
- the K-type CpG ODN and poly dA may be directly linked by a covalent bond or may be linked via a spacer sequence.
- spacer sequence is meant a nucleotide sequence comprising one or more nucleotides inserted between two adjacent components.
- the length of the spacer sequence is such that the complex of the present invention has an immunostimulatory activity (preferably an activity to activate B cells to produce IL-6 and an activity to activate dendritic cells to produce IFN- ⁇ ).
- an immunostimulatory activity preferably an activity to activate B cells to produce IL-6 and an activity to activate dendritic cells to produce IFN- ⁇ .
- it is usually 1 to 10 nucleotides long, preferably 1 to 5 nucleotides long, more preferably 1 to 3 nucleotides long.
- K-type CpG ODN and poly dA are linked by a direct covalent bond.
- the oligodeoxynucleotide of the present invention may have an additional nucleotide sequence at its 5 'end and / or 3' end in addition to K-type CpG ODN, poly dA and an optional spacer sequence.
- the length of the additional nucleotide sequence indicates that the complex of the present invention has an immunostimulatory activity (preferably an activity to activate B cells to produce IL-6, and a dendritic cell to produce IFN- ⁇ .
- the length is usually 1 to 10 nucleotides, preferably 1 to 5 nucleotides, more preferably 1 to 3 nucleotides.
- the oligodeoxynucleotide of the present invention does not contain such additional nucleotide sequences at the 5 'end and / or the 3' end. That is, the oligodeoxynucleotide of the present invention preferably comprises K-type CpG ODN, poly dA and an optional spacer sequence, and more preferably comprises K-type CpG ODN and poly dA.
- the oligodeoxynucleotide of the present invention comprises K-type CpG ODN (specifically, for example, an oligodeoxynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1) and poly dA, and comprises K-type CpG ODN. Is located at the 5 ′ end of the oligodeoxynucleotide and poly dA at the 3 ′ end.
- the oligodeoxynucleotide comprising the nucleotide sequence represented by SEQ ID NO: 1 has a length of 20 to 60 nucleotides (more preferably 30 to 50 nucleotides (30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50 nucleotides long), most preferably 30-45 nucleotides long (30, 31, 32) , 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45 nucleotides))), which is an oligodeoxynucleotide bound to, for example, SEQ ID NO: 2 or 9 It is an oligodeoxynucleotide consisting of the nucleotide sequence represented by ⁇ 12.
- the total length of the oligodeoxynucleotide of the present invention is generally 30 to 200 nucleotides, preferably 35 to 100 nucleotides, more preferably 40 to 80 nucleotides (specifically, 40, 41, 42, 43, 44, 45). , 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70 71, 72, 73, 74, 75, 76, 77, 78, 79 or 80 nucleotides long), more preferably 50 to 70 nucleotides long (specifically, 50, 51, 52, 53, 54, 55).
- the oligodeoxynucleotide of the present invention may be appropriately modified so as to be resistant to in vivo degradation (eg, degradation by exo or endonuclease).
- the modification comprises a phosphorothioate modification or a phosphorodithioate modification. That is, part or all of the phosphodiester bond in the oligodeoxynucleotide of the present invention is substituted by a phosphorothioate bond or a phosphorodithioate bond.
- the oligodeoxynucleotide of the present invention comprises a modification of a phosphodiester bond, more preferably the modification of a phosphodiester bond is a non-phosphorothioate bond (ie, as described in WO 95/26204).
- a modification of a phosphodiester bond is a non-phosphorothioate bond (ie, as described in WO 95/26204).
- One of the bridging oxygen atoms is replaced by a sulfur atom). That is, part or all of the phosphodiester bond in the oligodeoxynucleotide of the present invention is replaced by a phosphorothioate bond.
- the oligodeoxynucleotide of the present invention preferably contains a modification by phosphorothioate linkage or phosphorodithioate linkage in K-type CpG ODN, more preferably all of the phosphodiester linkage of K-type CpG ODN is Substituted with a phosphorothioate linkage.
- the oligodeoxynucleotide of the present invention preferably contains a phosphorothioate bond or a phosphorodithioate bond in poly dA, and more preferably all of the phosphodiester bonds of poly dA are replaced with phosphorothioate bonds. Is done.
- oligodeoxynucleotides including humanized K-type CpG oligodeoxynucleotides and polydeoxyadenylates of the invention, are replaced with phosphorothioate linkages.
- the oligodeoxynucleotide of the present invention has a length of 20 to 60 nucleotides (more preferably 30 to 50 nucleotides (30, 30) at the 3 ′ end of a humanized K-type CpG oligodeoxynucleotide (eg, SEQ ID NO: 1).
- the phosphorothioate bond in the oligodeoxynucleotide of the present invention, not only the resistance to degradation but also the immunostimulatory activity (for example, the activity of activating pDC to produce IFN- ⁇ ), and CpG- ⁇ -1, This is because high yield of 3-glucan complex and enhancement of anticancer activity are expected.
- the phosphorothioate bond is synonymous with the phosphorothioate skeleton
- the phosphate diester bond is synonymous with the phosphate skeleton.
- the oligodeoxynucleotides of the present invention include any pharmaceutically acceptable salts, esters, or salts of such esters of the above oligodeoxynucleotides.
- the pharmaceutically acceptable salts of the oligodeoxynucleotide of the present invention are preferably alkali metal salts such as sodium salt, potassium salt and lithium salt, alkaline earth metal salts such as calcium salt and magnesium salt, aluminum Metal salts such as salts, iron salts, zinc salts, copper salts, nickel salts, cobalt salts; inorganic salts such as ammonium salts, t-octylamine salts, dibenzylamine salts, morpholine salts, glucosamine salts, phenylglycine alkyl esters Salt, ethylenediamine salt, N-methylglucamine salt, guanidine salt, diethylamine salt, triethylamine salt, dicyclohexylamine salt, N, N'-dibenzylethylenediamine salt, chloroprocaine salt, procaine salt, diethanolamine salt, N-benzyl-phenethylamine Salt, piperazine salt Amine salts such as
- the oligodeoxynucleotide of the present invention may be in any form of single strand, double strand, and triple strand, but is preferably single strand.
- the oligodeoxynucleotide of the present invention is preferably isolated. “Isolated” means that an operation to remove factors other than the target component has been performed, and that the naturally occurring state has been removed.
- the purity of the “isolated oligodeoxynucleotide” (percentage of the desired oligodeoxynucleotide weight in the total weight of the evaluation object) is usually 70% or more, preferably 80% or more, more preferably 90% or more, More preferably, it is 99% or more.
- the oligodeoxynucleotide of the present invention has excellent immunostimulatory activity (for example, the activity of activating B cells (preferably human B cells) to produce IL-6), and thus is useful as an immunostimulator or the like. . Furthermore, since the oligodeoxynucleotide of the present invention has a property of forming a triple helix structure with two ⁇ -1,3-glucans (preferably, schizophyllan, lentinan or scleroglucan), the complex of the present invention Useful for preparation.
- the present invention provides a complex containing the oligodeoxynucleotide of the present invention and ⁇ -1,3-glucan (hereinafter referred to as the complex of the present invention).
- the above-mentioned oligodeoxynucleotide of the present invention contains K-type CpG ODN, it alone activates an immunostimulatory activity peculiar to K-type CpG ODN (for example, B cells (preferably human B cells) and IL -6), and immunostimulatory activity peculiar to D-type CpG ODN (for example, the activity of activating plasmacytoid dendritic cells to produce IFN- ⁇ ) is poor.
- K-type CpG ODN for example, B cells (preferably human B cells) and IL -6
- D-type CpG ODN for example, the activity of activating plasmacytoid dendritic cells to produce IFN- ⁇
- ⁇ -1,3-glucan preferably lentinan, schizophyllan
- an activation activity for example, an activity of activating plasmacytoid dendritic cells to produce IFN- ⁇
- the complex of the present invention has an immunostimulatory activity peculiar to K-type CpG ODN (for example, an activity that activates B cells (preferably human B cells) to produce IL-6) and D-type CpG ODN.
- Specific immunostimulatory activity for example, the activity of activating plasmacytoid dendritic cells (preferably human plasmacytoid dendritic cells) to produce IFN- ⁇ ).
- ⁇ -1,3-glucan used in the present invention include schizophyllan, scleroglucan, curdlan, parkan, glyphoran, lentinan, laminaran and the like.
- the ⁇ -1,3-glucan preferably contains a large amount of 1,6-glucopyranoside branches (side chain ratio 33 to 40%), such as schizophyllan, lentinan or scleroglucan. More preferred is schizophyllan.
- Lentinan is a known ⁇ -1,3-1,6-glucan derived from Shiitake mushroom, has a molecular formula of (C6H1005) n, and a molecular weight of about 300,000 to 700,000. It hardly dissolves in water, methanol, ethanol (95), or acetone, but dissolves in DMSO or sodium hydroxide aqueous solution, which are polar organic solvents.
- Lentinan has an effect of enhancing activated macrophages, killer T cells, natural killer cells and antibody-dependent macrophage-mediated cytotoxicity (ADMC) activity (Hamuro, J., et al .: Immunology, 39, 551-559, 1980, Hamuro, J., et al .: Int. J. Immunopharmacol., 2, 171, 1980, Herlyn, D., et al .: Gann, 76, 37-42, 1985).
- ADMC antibody-dependent macrophage-mediated cytotoxicity
- Schizophyllan is a known soluble ⁇ -glucan derived from Shirohirotake.
- SPG consists of a ⁇ - (1 ⁇ 3) -D-glucan main chain and one ⁇ - (1 ⁇ 6) -D-glucosyl side chain for each three glucoses (Tabata, K., Ito). , W., Kojima, T., Kawabata, S. and Misaki A., “Carbohydr. Res.”, 1981, 89, 1, p.
- SPG has been used for more than 20 years as a clinical drug for intramuscular injection of immunity enhancement method against gynecological cancer (Shimizu, Chen, Kazumi, Masumi, “Biotherapy”, 1990, 4, p.
- complex refers to a product obtained by association of a plurality of molecules through non-covalent or covalent bonds such as electrostatic bonds, van der Waals bonds, hydrogen bonds, and hydrophobic interactions. Means.
- the complex of the present invention preferably has a triple helical structure.
- two of the three chains forming the triple helix structure are ⁇ -1,3-glucan chains, and one is the polydeoxyadenylic acid in the oligodeoxynucleotide of the present invention. Is a chain.
- the complex may partially include a portion that does not form a triple helical structure.
- composition ratio of oligodeoxynucleotide and ⁇ -1,3-glucan in the complex of the present invention depends on the chain length of polydeoxyadenylic acid in the oligodeoxynucleotide, the length of ⁇ -1,3-glucan, etc. Can change. For example, when the lengths of ⁇ -1,3-glucan chains and polydeoxyadenylic acid chains are equivalent, two ⁇ -1,3-glucan chains and one oligodeoxynucleotide of the present invention Can associate to form a triple helix structure.
- the complex of the present invention is a complex containing humanized K-type CpG ODN and ⁇ -1,3-glucan (eg, lentinan, schizophyllan, scleroglucan, curdlan, perchiman, glyphoran, laminaran), preferably Is a complex consisting of humanized K-type CpG ODN and ⁇ -1,3-glucan (eg, lentinan, schizophyllan, scleroglucan).
- humanized K-type CpG ODN and ⁇ -1,3-glucan eg, lentinan, schizophyllan, scleroglucan
- the oligodeoxynucleotide consisting of the nucleotide sequence represented by SEQ ID NO: 1 has a length of 20 to 60 nucleotides on the 3 ′ side (specifically, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59 or 60 nucleotides in length), and oligodeoxynucleotides in which all of the phosphodiester bonds are replaced with phosphorothioate bonds, and ⁇ -1, A complex composed of 3-glucan (eg, lentinan, schizophyllan) (eg, K3-dA20-60-LNT, K3-dA20-60-SPG);
- a complex (eg, K3-dA30-50-LNT, K3-dA30-50-SPG) composed of an oligodeoxynucleotide and ⁇ -1,3-glucan (eg, lentinan, schizophyllan), most preferably a sequence
- the oligodeoxynucleotide consisting of the nucleotide sequence represented by No.
- K3-dA30-45-LNT, K3-dA30-45-SPG consisting of an oligodeoxynucleotide substituted with a phosphorothioate bond and a ⁇ -1,3-glucan (eg, lentinan, schizophyllan) is there.
- the method for preparing the complex of the present invention can be carried out in the same manner as the conditions described in Non-Patent Documents 21 to 24 and Japanese Patent Application Laid-Open No. 2008-1000091. That is, ⁇ -1,3-glucan, which originally exists as a triple helical structure in nature, is converted into an aprotic organic polar solvent (dimethyl sulfoxide (DMSO), acetonitrile, acetone, etc.) or an alkaline aqueous solution (sodium hydroxide, potassium hydroxide).
- DMSO dimethyl sulfoxide
- acetonitrile acetone
- alkaline aqueous solution sodium hydroxide, potassium hydroxide
- the solution of the single-stranded ⁇ -1,3-glucan thus obtained and the oligodeoxynucleotide solution of the present invention aqueous solution, buffer solution having a pH near neutral, or acidic buffer solution, preferably Aqueous solution or a buffered aqueous solution having a pH near neutral
- the complex of the present invention is formed by the two ⁇ -1,3-glucan chains and the poly dA chain in the oligodeoxynucleotide forming a triple helical structure.
- the complex By performing purification by size exclusion chromatography, ultrafiltration, dialysis and the like on the produced complex, oligodeoxynucleotides not formed in the complex can be removed. Further, by performing purification by anion exchange chromatography on the produced complex, ⁇ -1,3-glucan having no complex formed can be removed. By the above method, the complex can be appropriately purified.
- the formation of the complex of the present invention is performed by measuring, for example, conformational change by CD (circular dichroism) spectrum, UV absorption shift by size exclusion chromatography, gel electrophoresis, microchip electrophoresis, capillary electrophoresis. Although it can confirm, it is not restricted to this.
- CD circular dichroism
- the mixing ratio of the oligodeoxynucleotide of the present invention and ⁇ -1,3-glucan can be appropriately set in consideration of the length of the poly dA chain and the like, but usually the molar ratio (SPG / ODN) is 0. It is 02 to 2.0, preferably 0.1 to 0.5. In a further embodiment, the molar ratio ( ⁇ -1,3-glucan (LNT, etc.) / ODN) is, for example, 0.005 to 1.0, preferably 0.020 to 0.25.
- LNT is dissolved in 0.05 to 2N, preferably 0.1 to 1.5N alkaline aqueous solution (for example, 0.25N sodium hydroxide aqueous solution) and left at 1 ° C to 40 ° C for 10 hours to 4 days (for example, Left overnight at room temperature) to prepare a single-stranded LNT aqueous solution (eg, 50 mg / ml LNT aqueous solution).
- alkaline aqueous solution for example, 0.25N sodium hydroxide aqueous solution
- 4 days for example, Left overnight at room temperature
- the LNT aqueous solution and a separately prepared CpG aqueous solution are mixed at a molar ratio (LNT / ODN) of 0.005 to 1.0, and then an acidic buffered aqueous solution (for example, NaH2PO4) is added to neutralize and maintained at 1-40 ° C. for 6 hours to 4 days (eg, overnight at 4 ° C.) to complete the complexation.
- an acidic buffered aqueous solution for example, NaH2PO4
- the composite of the present invention takes the form of bowl-like particles.
- the particle diameter is the same as the diameter of a particle formed by ⁇ -1,3-glucan (eg, schizophyllan) used as a material naturally having a triple helical structure, and the average particle diameter is usually 10-100 nm, preferably 20-50 nm.
- the particle diameter can be measured by a dynamic light scattering method at 80 ° C. using a Malvern Instruments Zeta Sizer after dissolving the complex in water.
- the complex of the present invention is preferably isolated.
- the purity of the “isolated complex” (percentage of the target complex weight in the total weight of the object to be evaluated) is usually 70% or more, preferably 80% or more, more preferably 90% or more, still more preferably Is 99% or more.
- the complex of the present invention has excellent immunostimulatory activity in addition to anticancer activity, and in particular, immunostimulatory activity peculiar to K-type CpG ODN (for example, B cells (preferably human B cells)) and the immunostimulatory activity peculiar to D-type CpG ODN (eg, plasmacytoid dendritic cells (preferably human plasmacytoid dendritic cells) are activated to activate IFN- Since it has both of (activity which produces (alpha)), since an effect can be provided also as an immunostimulant etc., it may be advantageous.
- immunostimulatory activity peculiar to K-type CpG ODN for example, B cells (preferably human B cells)
- D-type CpG ODN eg, plasmacytoid dendritic cells (preferably human plasmacytoid dendritic cells) are activated to activate IFN- Since it has both of (activity which produces (alpha)), since an effect can be provided also as an immunostimulant etc., it may be advantageous.
- a K-type CpG ODN for example, a complex containing SEQ ID NO: 2, 11, 12 and SPG and a complex containing a K-type CpG ODN (for example, SEQ ID NO: 2) and SPG (K3-SPG) induces an inflammatory response.
- Potency pan-IFN-a, IL-6, etc.
- serum antigen-specific IgG antibody titer total IgG, IgG2c, etc.
- IFN- antigen-specific cytokine production ability
- the present invention provides a pharmaceutical composition comprising the oligodeoxynucleotide of the present invention or the complex of the present invention.
- the pharmaceutical composition of the present invention can be obtained by formulating the oligodeoxynucleotide of the present invention or the complex of the present invention according to conventional means.
- the pharmaceutical composition of the present invention comprises the oligodeoxynucleotide or complex of the present invention and a pharmacologically acceptable carrier.
- the pharmaceutical composition may further contain an antigen.
- Such a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
- injections are dosage forms such as intravenous injections, subcutaneous injections, intradermal injections, intramuscular injections, infusions, and the like. May be included.
- Such an injection can be prepared according to a known method.
- a method for preparing an injection it can be prepared, for example, by dissolving or suspending the oligodeoxynucleotide or complex of the present invention in a sterile aqueous solvent usually used for injection.
- the aqueous solvent for injection include distilled water; physiological saline; phosphate buffer, carbonate buffer, Tris buffer, acetate buffer, and other buffer solutions.
- the pH of such an aqueous solvent is 5 to 10, preferably 6 to 8.
- the prepared injection solution is preferably filled in a suitable ampoule.
- a powder formulation of the oligodeoxynucleotide or complex of the present invention can also be prepared by subjecting the suspension of the oligodeoxynucleotide or complex of the present invention to a treatment such as vacuum drying or freeze drying.
- the oligodeoxynucleotide or complex of the present invention can be stored in a powder state, and can be used by dispersing the powder with an aqueous solvent for injection at the time of use.
- the content of the oligodeoxynucleotide or complex of the present invention in the pharmaceutical composition is usually about 0.1 to 100% by weight, preferably about 1 to 99% by weight, more preferably about 10 to 10% by weight of the whole pharmaceutical composition. About 90% by weight.
- the pharmaceutical composition of the present invention may contain the oligodeoxynucleotide or complex of the present invention alone as an active ingredient, and contains the oligodeoxynucleotide or complex of the present invention in combination with other active ingredients. It may be.
- the oligodeoxynucleotides and conjugates of the present invention have been found to have anticancer activity alone. Such an effect can be said to be an unexpected effect from the characteristics of the present invention that have been developed as an adjuvant. Therefore, it can be used as an adjuvant so far, that is, it is not necessary to administer with a cancer antigen, and it is not limited to a specific cancer type.
- An anticancer agent that acts mildly on the body as an agent will be provided.
- it since it also has immunostimulatory activity, it can also be expected to have immune activation activity against other diseases, and to have a synergistic effect on cancer patients with weak physical strength.
- the present invention has excellent immunostimulatory activity, so the oligodeoxynucleotide, complex and pharmaceutical composition of the present invention can be used as an immunostimulator.
- a mammal a primate such as a human, a rodent such as a mouse
- an immune reaction in the mammal can be induced.
- the complex of the present invention has the activity characteristics of D-type CpG ODN and stimulates peripheral blood mononuclear cells to give type I interferons (Pan-IFN- ⁇ , IFN- ⁇ 2, etc.) and type II interferons ( Since both IFN- ⁇ ) are produced in large quantities, they are useful as type I interferon production inducers, type II interferon production inducers, type I and type II interferon production inducers. Since it induces the production of both type I and type II interferons, the complex of the present invention and the pharmaceutical composition containing the same prevent or prevent diseases in which one or both of type I and type II interferons are effective. Useful for treatment.
- Cytotoxic T lymphocytes in a subject who has received the administration can be antigen-specifically activated to prevent or treat cancer directly (as a single agent effect).
- the “subject (person)” refers to a subject to be diagnosed or detected or treated according to the present invention (for example, an organism such as a human or a cell, blood, serum, etc. removed from the organism). .
- drug drug
- drug may also be a substance or other element (eg energy such as light, radioactivity, heat, electricity).
- Such substances include, for example, proteins, polypeptides, oligopeptides, peptides, polynucleotides, oligonucleotides, nucleotides, nucleic acids (eg, DNA such as cDNA, genomic DNA, RNA such as mRNA), poly Saccharides, oligosaccharides, lipids, small organic molecules (for example, hormones, ligands, signaling substances, small organic molecules, molecules synthesized by combinatorial chemistry, small molecules that can be used as pharmaceuticals (for example, small molecule ligands, etc.)) , These complex molecules are included, but not limited thereto.
- a polynucleotide having a certain sequence homology to the sequence of the polynucleotide (for example, 70% or more sequence identity) and complementarity examples include, but are not limited to, a polypeptide such as a transcription factor that binds to the promoter region.
- Factors specific for a polypeptide typically include an antibody specifically directed against the polypeptide or a derivative or analog thereof (eg, a single chain antibody), and the polypeptide is a receptor.
- specific ligands or receptors in the case of ligands, and substrates thereof when the polypeptide is an enzyme include, but are not limited to.
- treatment refers to prevention of worsening of a disease or disorder when a disease or disorder (eg, cancer, allergy) occurs, preferably, maintaining the status quo. More preferably, it means reduction, further preferably elimination, which includes the ability to exert a symptom improving effect or a preventive effect on one or more symptoms associated with a patient's disease or disease. Diagnosing in advance and performing appropriate treatment is referred to as “companion treatment”, and the diagnostic agent therefor is sometimes referred to as “companion diagnostic agent”.
- the term “therapeutic agent (agent)” refers to any drug that can treat a target condition (for example, diseases such as cancer, allergy, etc.) in a broad sense.
- the “therapeutic agent” may be a pharmaceutical composition comprising an active ingredient and one or more pharmacologically acceptable carriers.
- the pharmaceutical composition can be produced by any method known in the technical field of pharmaceutics, for example, by mixing the active ingredient and the carrier.
- the form of use of the therapeutic agent is not limited as long as it is a substance used for treatment, and it may be an active ingredient alone or a mixture of an active ingredient and an arbitrary ingredient.
- the shape of the carrier is not particularly limited, and may be, for example, a solid or a liquid (for example, a buffer solution).
- the therapeutic agent for cancer, allergy and the like includes a drug (preventive agent) used for preventing cancer, allergy and the like, or a suppressor for cancer, allergy and the like.
- prevention means that a certain disease or disorder (for example, allergy) is prevented from becoming such a state before it becomes such a state. Diagnosis can be performed using the drug of the present invention, and for example, allergies can be prevented using the drug of the present invention, or countermeasures for prevention can be taken as necessary.
- a certain disease or disorder for example, allergy
- prophylactic agent refers to any agent that can prevent a target condition (for example, a disease such as allergy) in a broad sense.
- the “kit” is a unit provided with a portion to be provided (eg, a test agent, a diagnostic agent, a therapeutic agent, an antibody, a label, an instruction, etc.) usually divided into two or more compartments.
- a portion to be provided eg, a test agent, a diagnostic agent, a therapeutic agent, an antibody, a label, an instruction, etc.
- This kit form is preferred when it is intended to provide a composition that should not be provided in admixture for stability or the like, but preferably used in admixture immediately before use.
- Such kits preferably include instructions or instructions that describe how to use the provided parts (eg, test agents, diagnostic agents, therapeutic agents, or how the reagents should be processed).
- the kit when the kit is used as a reagent kit, the kit usually contains instructions including usage of test agents, diagnostic agents, therapeutic agents, antibodies, etc. Is included.
- the “instruction sheet” describes the method for using the present invention for a doctor or other user.
- This instruction manual includes a word indicating that the detection method of the present invention, how to use a diagnostic agent, or administration of a medicine or the like is given.
- the instructions may include a word indicating that the administration site is oral or esophageal administration (for example, by injection).
- This instruction is prepared in accordance with the format prescribed by the national supervisory authority (for example, the Ministry of Health, Labor and Welfare in Japan and the Food and Drug Administration (FDA) in the United States, etc. in the United States) where the present invention is implemented, and is approved by the supervisory authority. It is clearly stated that it has been received.
- the instruction sheet is a so-called package insert and is usually provided as a paper medium, but is not limited thereto, and is in the form of, for example, an electronic medium (for example, a home page or e-mail provided on the Internet). But it can be provided.
- the invention provides an oligodeoxynucleotide comprising (a) a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide.
- an anticancer agent containing a complex comprising oligodeoxynucleotides arranged on the 3 ′ side and (b) ⁇ -1,3-glucan. In the present invention, it was found that the complex itself of the present invention acts as an anticancer agent.
- the anticancer agent of the present invention is administered without a cancer antigen.
- the anticancer agent of the present invention is administered so as to be delivered to the reticuloendothelial system and / or lymph nodes.
- the reticuloendothelial system and / or lymph nodes comprise tumors and phagocytic cells.
- the reticuloendothelial system includes the spleen and / or liver. Therefore, the anticancer agent of the present invention is characterized in that it is administered so as to be delivered to reticuloendothelial organs (spleen, liver, etc.) and / or lymph nodes including tumors and phagocytic cells.
- the complexes of the invention can be delivered to tumors and phagocytic cells, where cancer dead cells can be recruited to reticuloendothelial organs (spleen, liver, etc.). Indicated. Thereby, it is considered that cancer cells in the body can be further destroyed. Therefore, it can be said that the present invention can kill any cancer cell present in the body, not against a specific cancer using a specific cancer antigen as an adjuvant, It can be said that a remarkable effect is brought about in that an agent can be provided.
- the anticancer agent of the present invention is administered so as to be delivered to tumors and phagocytic cells without a cancer antigen.
- Such a delivery method may be any method, and for example, the administration may include systemic administration, but is not limited thereto.
- Systemic administration is preferred, and examples of systemic administration include intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, and intramuscular administration.
- the oligodeoxynucleotide used in the present invention is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4). , K3-dA25 (SEQ ID NO: 5), K3-dA30 (SEQ ID NO: 6), K3-dA35 (SEQ ID NO: 7), and the like.
- ⁇ -1,3-glucan used in the present invention may be schizophyllan (SPG), lentinan, scleroglucan, curdlan, perchiman, glyforan, laminaran and the like.
- SPG schizophyllan
- lentinan lentinan
- scleroglucan lentinan
- curdlan curdlan
- perchiman glyforan
- laminaran laminaran
- the complex of the present invention is K3-SPG or an analogue thereof.
- the class itself include, but are not limited to, those having a structure similar to K3 on the CpG side and those having a structure similar to SPG on the ⁇ -glucan side.
- the anti-cancer action is due to various mechanisms, it is not easy to conceive uses such as collecting dead cancer cells in the spleen. In particular, in systemic administration, the use for accumulating in a tumor and accumulating dead tumor cells in tissues such as the spleen is not conceivable.
- the expression of interleukin 12 (IL12) and / or interferon (IFN) ⁇ or its promoting effect is also due to a mechanism different from the anticancer action, and interleukin 12 (IL12) and / or interferon (IFN) ) Since the expression of ⁇ or its promotion can be exerted other than anti-cancer, it is not easy to come up with each other.
- each use of the CpG- ⁇ glucan complex of the present invention (anti-cancer use (single agent), use to accumulate cancer dead cells in the spleen, interleukin 12 (IL12) and / or interferon (IFN)) It can be said that there is a relationship that cannot be easily conceived with each other.
- the present invention provides an oligodeoxynucleotide comprising (a) a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide.
- the complex of the present invention can accumulate cancer dead cells in the reticuloendothelial system (including spleen and / or liver) and / or lymph nodes. It was.
- treatment with a complex of the invention such as K3-SPG has been demonstrated to induce tumor cell death in a manner that is dependent on both IL12p40 and IFN-I.
- CpG is targeted to phagocytic cells in the tumor microenvironment.
- cancer dead cells Once cancer dead cells accumulate in the reticuloendothelial system (including spleen and / or liver) and / or lymph nodes, the released tumor dead cells then induce anti-tumor CTL against multiple tumor antigens.
- the cancer cells in the body can be killed and cured as if they were attacked with a shotgun. Without wishing to be bound by theory, it is important, although not necessarily essential for K3-SPG monotherapy, to produce both IL12 and IFN-I cytokines in the tumor microenvironment.
- the oligodeoxynucleotide used in the present invention is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4). , K3-dA25 (SEQ ID NO: 5), K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- the ⁇ -1,3-glucan used in the present invention is selected from the group consisting of schizophyllan (SPG), scleroglucan, curdlan, parkan, glyphoran and laminaran.
- the complex of the present invention is K3-SPG.
- the reticuloendothelial system and / or lymph nodes targeted by the composition of the present invention comprises tumors and phagocytic cells.
- the reticuloendothelial system includes the spleen and / or liver.
- the composition of the present invention is characterized in that it is administered so as to be delivered to reticuloendothelial organs (spleen, liver, etc.) and / or lymph nodes containing tumors and phagocytic cells.
- the complexes of the invention can be delivered to tumors and phagocytic cells, where cancer dead cells can be recruited to reticuloendothelial organs (spleen, liver, etc.). Indicated. Thereby, it is considered that cancer cells in the body can be further destroyed. Therefore, it can be said that the present invention can kill any cancer cell present in the body, not against a specific cancer using a specific cancer antigen as an adjuvant, It can be said that a remarkable effect is brought about in that an agent can be provided.
- the anticancer agent of the present invention is administered so as to be delivered to tumors and phagocytic cells without a cancer antigen.
- Such a delivery method may be any method, and for example, the administration may include systemic administration, but is not limited thereto.
- Systemic administration is preferred, and examples of systemic administration include intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, and intramuscular administration.
- the present invention provides (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide.
- an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide.
- IFN interferon
- K3-SPG monotherapy such as acting as an anti-cancer agent and other It is also important in applications.
- examples of such treatment include, but are not limited to, cancer, chronic infectious diseases such as viruses, and prevention of viral infection.
- the oligodeoxynucleotide used in the present invention is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4). , K3-dA25 (SEQ ID NO: 5), K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- the ⁇ -1,3-glucan used in the present invention is selected from the group consisting of schizophyllan (SPG), scleroglucan, curdlan, parkan, glyphoran and laminaran.
- the complex of the present invention is K3-SPG.
- the present invention is provided as the above-mentioned various forms of medicaments (therapeutic or preventive).
- the administration route of the therapeutic agent is preferably one that is effective in the treatment, and may be, for example, intravenous, subcutaneous, intramuscular, intraperitoneal, or oral administration.
- the administration form may be, for example, an injection, capsule, tablet, granule or the like.
- Aqueous solutions for injection may be stored, for example, in vials or stainless steel containers.
- the aqueous solution for injection may contain, for example, physiological saline, sugar (for example, trehalose), NaCl, or NaOH.
- the therapeutic agent may contain, for example, a buffer (for example, phosphate buffer), a stabilizer and the like.
- compositions, medicaments, therapeutic agents, prophylactic agents, etc. of the present invention comprise a therapeutically effective amount of a therapeutic agent or active ingredient, and a pharmaceutically acceptable carrier or excipient.
- pharmaceutically acceptable refers to a licensed or otherwise recognized pharmacopoeia of a government for use in animals, and more particularly in humans, by a government supervisory authority. It means that it is enumerated.
- carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic agent is administered.
- Such carriers can be sterile liquids, such as water and oils, including but not limited to those of petroleum, animal, vegetable or synthetic origin, including but not limited to peanut oil, soybean oil, minerals Oil, sesame oil, etc. are included.
- Water is a preferred carrier when the drug is administered orally.
- Saline and aqueous dextrose are preferred carriers when the pharmaceutical composition is administered intravenously.
- saline solutions and aqueous dextrose and glycerol solutions are used as liquid carriers for injectable solutions.
- Suitable excipients include light anhydrous silicic acid, crystalline cellulose, mannitol, starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, chloride Sodium, nonfat dry milk, glycerol, propylene, glycol, water, ethanol, carmellose calcium, carmellose sodium, hydroxypropylcellulose, hydroxypropylmethylcellulose, polyvinylacetal diethylaminoacetate, polyvinylpyrrolidone, gelatin, medium chain fatty acid triglyceride, polyoxyethylene hardening Castor oil 60, sucrose, carboxymethylcellulose, corn starch, inorganic salts and the like are included.
- compositions can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents, if desired.
- These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. It is also possible to formulate the composition as a suppository, with traditional binders and carriers such as triglycerides. Oral formulations may also include standard carriers such as pharmaceutical grade mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate. Examples of suitable carriers are E.I. W. Martin, Remington's Pharmaceutical Sciences (Mark Publishing Company, Easton, USA).
- compositions contain a therapeutically effective amount of the therapeutic agent, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
- the formulation must be suitable for the mode of administration.
- surfactants, excipients, coloring agents, flavoring agents, preservatives, stabilizers, buffering agents, suspending agents, tonicity agents, binders, disintegrating agents, lubricants, fluidity Accelerators, flavoring agents and the like may be included.
- the “salt” includes, for example, an anion salt formed with any acidic (eg, carboxyl) group or a cation salt formed with any basic (eg, amino) group.
- Salts include inorganic salts or organic salts, for example, Berge et al. , J .; Pharm. Sci. , 1977, 66, 1-19. Examples thereof include metal salts, ammonium salts, salts with organic bases, salts with inorganic acids, salts with organic acids, and the like.
- a “solvate” is a compound formed by a solute and a solvent. For solvates, see, for example, J. Org. Honiget al.
- the solvent is water, the solvate formed is a hydrate. This solvent is preferably one that does not interfere with the biological activity of the solute. Examples of such preferred solvents include, but are not limited to, water or various buffers.
- “chemical modification” includes, for example, modification with PEG or a derivative thereof, fluorescein modification, biotin modification, or the like.
- various delivery systems are known, and such systems can be used to administer the therapeutic agent of the present invention to an appropriate site (eg, esophagus).
- Such systems include, for example, encapsulation in liposomes, microparticles, and microcapsules: the use of recombinant cells capable of expressing therapeutic agents (eg, polypeptides), receptor-mediated endocytosis Use; such as the construction of therapeutic nucleic acids as part of a retroviral vector or other vector.
- Introduction methods include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
- an inhaler or nebulizer can be used with an aerosolizing agent and can be administered with other biologically active agents. Administration can be systemic or local.
- the present invention can be administered by any appropriate route such as direct injection into cancer (lesion).
- the composition can be formulated as a pharmaceutical composition adapted for human administration according to known methods. Such compositions can be administered by injection. Typically, compositions for injection administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition can also include a solubilizing agent and a local anesthetic such as lidocaine to ease pain at the site of the injection. In general, the ingredients are supplied separately or mixed together in a unit dosage form, for example in a sealed container such as an ampoule or sachet indicating the amount of active agent, lyophilized powder or water-free concentration Can be supplied as a product.
- a sealed container such as an ampoule or sachet indicating the amount of active agent, lyophilized powder or water-free concentration Can be supplied as a product.
- composition is to be administered by infusion
- it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
- an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
- compositions, medicament, therapeutic agent, and preventive agent of the present invention in a neutral form, salt form, or other prodrug (for example, ester).
- pharmaceutically acceptable salts include those formed with free carboxyl groups derived from hydrochloric acid, phosphoric acid, acetic acid, oxalic acid, tartaric acid, isopropylamine, triethylamine, 2-ethylaminoethanol, histidine, procaine And those formed with free amine groups such as those derived from, and those derived from sodium, potassium, ammonium, calcium, ferric hydroxide, and the like.
- the amount of the therapeutic agent of the invention effective for the treatment of a particular disorder or condition can vary depending on the nature of the disorder or condition, but can be determined by those skilled in the art by standard clinical techniques based on the description herein. Furthermore, in some cases, in vitro assays can be used to help identify optimal dosage ranges.
- the exact dose to be used in the formulation can also vary depending on the route of administration and the severity of the disease or disorder and should be determined according to the judgment of the attending physician and the circumstances of each patient. However, the dose is not particularly limited, and may be, for example, 0.001, 1, 5, 10, 15, 100, or 1000 mg / kg body weight per dose, and within the range of any two of these values There may be.
- the dosing interval is not particularly limited.
- the dose, administration interval, and administration method may be appropriately selected depending on the age, weight, symptoms, target organ, etc. of the patient.
- the therapeutic agent preferably contains a therapeutically effective amount or an effective amount of an active ingredient that exhibits a desired action. If the malignant tumor marker is significantly decreased after administration, it may be determined that there is a therapeutic effect. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.
- a “patient” or “subject” is a human or non-human mammal (eg, mouse, guinea pig, hamster, rat, mouse, rabbit, pig, sheep, goat, cow, horse, One or more of a cat, dog, marmoset, monkey, or chimpanzee).
- a human or non-human mammal eg, mouse, guinea pig, hamster, rat, mouse, rabbit, pig, sheep, goat, cow, horse, One or more of a cat, dog, marmoset, monkey, or chimpanzee.
- the pharmaceutical composition or therapeutic agent or prophylactic agent of the present invention can be provided as a kit.
- the present invention provides a drug pack or kit comprising one or more containers filled with one or more components of the composition or medicament of the present invention.
- a drug pack or kit comprising one or more containers filled with one or more components of the composition or medicament of the present invention.
- associated with such containers manufactured, used or sold for human administration by a government agency in a manner prescribed by the government agency that regulates the manufacture, use or sale of a pharmaceutical or biological product. It is also possible to indicate information indicating authorization.
- a pharmaceutical composition comprising an ingredient of the present invention can be administered via liposomes, microparticles, or microcapsules.
- the formulation procedure as a medicine such as a therapeutic drug and a preventive drug of the present invention is known in the art, and is described in, for example, the Japanese Pharmacopoeia, the US Pharmacopoeia, and the pharmacopoeia of other countries. Accordingly, those skilled in the art can determine the embodiment, such as the amount to be used, without undue experimentation as described herein.
- the oligonucleotide of the present invention can be synthesized by a standard method known in the art, for example, by using an automated DNA synthesizer (commercially available from Biosearch, Applied Biosystems, etc.). is there.
- an automated DNA synthesizer commercially available from Biosearch, Applied Biosystems, etc.
- it is possible to synthesize phosphorothioate oligonucleotides by the method of Stein et al. (Stein et al., 1988, Nucl. Acids Res. 16: 3209), and control pore glass polymer supports (Sarinet al., 1988, Proc. Natl. Acad. Sci. USA 85: 7448-7451), etc. can also be used to prepare methylphosphonate oligonucleotides.
- K3-dA40 SEQ ID NO: 2
- K3-dA35 SEQ ID NO: 7
- K3-dA30 SEQ ID NO: 6
- K3-dA25 SEQ ID NO: 5
- K3-dA20 SEQ ID NO: 4
- This oligodeoxynucleotide can be obtained by using the conventional solid phase phosphoramidite method (Nucleic Acids in Chemistry and Biology, 3. Chemical synthesis (1990) ed. G. Michael Blackburn and Michael J. GaitOx. Synthesized.
- Ovalbumin was purchased from Seikagaku Corporation. DQ-OVA, Alexa488-OVA, CFSE, and Lipofectamine 2000 were purchased from Invitrogen. Hoechst 33258, zymozan and curdlan were purchased from SIGMA. Zymosan-Depleted was purchased from Invivogen. Clodronate liposomes were purchased from FormuMax. Influenza split product vaccine, formalin inactivated whole virus (WIV), and purified influenza virus (H1N1) were prepared as previously described (Koyama, S., et al., Science translational medicine 2, 25ra24 (2010) ).
- the formation of the complex was performed by using size exclusion chromatography and monitoring the shift of CpG ODN toward high molecular weight at 260 nm.
- System Agilent 1100 series, Column: Asahipak GF7M-HQ (Shodex) 2 ligation, Flow rate: 0.8 mL / min, Buffer: 10 mM EDTA PBS, pH 7.4, Temperature: 40 ° C.).
- EL4 and OVA expressing EL4 is a thymoma cell line of C57BL / 6J mice and was purchased from ATCC.
- B16 (melanoma) was purchased from the Japan Collection of Research Bioresources.
- B16F10 (melanoma) was purchased from Riken Cell Bank and MC38 (colon cancer) was Provided by Dr. JAMES Primus.
- Pan02 pancreatic cancer was purchased from Jackson's Laboratory.
- EL4, EG7, MC38, and Pan02 were cultured in complete RPMI (RPMI 1640 supplemented with 10% (v / v) fetal bovine serum (FBS), penicillin, and streptomycin).
- B16 and B16F10 were cultured in complete DMEM (DMEM supplemented with 10% (v / v) fetal bovine serum (FBS), penicillin, and streptomycin).
- Pan02 peritoneal seeding model In the Pan02 peritoneal seeding model, 1 ⁇ 10 6 Pan02 cells (100 ⁇ l at 1 ⁇ 10 7 cells / mL in PBS) were injected intraperitoneally. CpG treatment was started 11 days after inoculation and all tumor nodules were excised from the peritoneum of mice on day 21 and their weight (g) was then measured. The dosage for CpG treatment is as described above.
- mice were treated with EG7 at day 0.
- c. Inoculate and receive PBS (control), Alexa 647-K3 (30 ⁇ g), or Alexa 647-K3-SPG (10 ⁇ g) i. v. Administered.
- PBS control
- Alexa 647-K3 30 ⁇ g
- Alexa 647-K3-SPG 10 ⁇ g
- mice were analyzed with IVIS® Lumina Imaging System and analysis software (Ver. 2.6, Xenogen), and images measured with relative fluorescence were measured in physical units (photons / photon / surface radiance). Second / cm 2 / sr).
- splenocytes were treated with K3-SPG on days 7, 9, 11 or untreated, C57BL / 6 mice carrying EG7 Or harvested on day 14 from Il12p40-Ifnar2 double knockout mice.
- erythrocytes were lysed with ACK lysis buffer (150 mM NH 4 Cl, 10 mM KHCO 3 , 0.1 mM Na 2 EDTA) and cells were maintained in complete RPMI.
- CD8 ⁇ + T cells were sorted by MACS (Miltenyi Biotec). CD8 ⁇ + T cells were sorted by the negative selection method. The sorted CD8 ⁇ + T cells were then stained with Xenolight DiR®.
- mice Stained CD8 ⁇ + T cells were injected or treated with recipient mice (EG7 on day 0, iv7 on days 0, and K3-SPG on days 7, 9, 11). C57BL / 6 mice or Il12p40-Ifnar2 double knockout mice). Twenty-four hours after transferring the stained cells, the mice were analyzed with the IVIS® Lumina Imaging System (Ver. 2.6). The region of interest was aggregated into the tumor region and the fluorescence intensity was analyzed with Living Image Software (Ver. 2.6, Xenogen).
- C57BL / 6J mice (6-8 weeks old, female, CLEA Japan) were treated with Alexa 647-K3 (30 ⁇ g), Alexa 647-K3-SPG (10 ⁇ g), and dextran-PE (20 ⁇ g) i.e. from the tail vein.
- Splenocytes were injected i.p. with K3-SPG on days 7, 9, 11. v. Recovered on day 14 from treated or untreated C57BL / 6 mice carrying EG7 or Il12p40-Ifnar2 double knockout mice. After preparation of splenocytes, erythrocytes were lysed with ACK lysis buffer and the cells were maintained with complete RPMI. Splenocytes were isolated from H-2K b OVA tetramer (MBL), anti-CD8 ⁇ antibody (KT15), anti-TCR ⁇ antibody (H57-597), anti-CD62L antibody (MEL-14), and anti-CD44 antibody (IM7), and 7-amino.
- MBL H-2K b OVA tetramer
- KT15 anti-CD8 ⁇ antibody
- KT15 anti-TCR ⁇ antibody
- MEL-14 anti-CD44 antibody
- IM7 anti-CD44 antibody
- Splenocytes were injected i.p. with K3-SPG on days 7, 9, 11. v. Harvested at day 12 from treated or untreated C57BL / 6 mice carrying EG7 or Il12p40-Ifnar2 double knockout mice. After preparation of splenocytes, erythrocytes were lysed with ACK lysis buffer and the cells were maintained in complete RPMI. Splenocytes were stained with anti-CD45 antibody (APC) and the number of CD45 ⁇ cells was determined by flow cytometry.
- APC anti-CD45 antibody
- CD45 ⁇ cells were sorted by INFLUX (BD Bioscience) from tumor bearing C57BL / 6 mice treated with K3-SPG.
- mice C57BL / 6 mice were challenged with 5 ⁇ 10 5 CD45 ⁇ cells on day ⁇ 7. v. Administered. Seven days after immunization, mice were challenged with 5 ⁇ 10 5 EG7 cells on day 0. c. Vaccinated.
- Mouse IL-12p40, mouse IL-13, and human IFN ⁇ levels were measured using an R & D ELISA kit.
- Example 1 Intravenous injection of K3-SPG induces strong tumor growth inhibition without any additional tumor antigen
- mice were inoculated with EG7 (OVA expressing mouse thymoma cell line) on the right flank on day 0, administered subcutaneously (id) near the tail base, intratumoral (it), Or three times with PBS and equimolar amounts of K3 (30 ⁇ g) or K3-SPG (10 ⁇ g) via three different routes of intravenous (iv) administration (7, 9, 11 of inoculation) days after). Tumor size was measured every 2-3 days until day 23.
- K3-SPG forms nanoparticles with a size of about 30 nm (Kobiyama, K., et al. Proc. Natl. Acad. Sci. USA 111, 3086-3091 (2014)).
- IVIS imaging uses K3-SPG instead of K3, i. v. It was revealed that it accumulated at the tumor site 1 hour after administration (FIG. 4a). It was observed that the accumulation of K3-SPG in the tumor is well related to the effectiveness of CpG monotherapy treatment for tumor shrinkage (FIG. 2).
- IHC immunohistochemistry
- EG7 cells express CD3e on their surface (because EG7 is derived from a thymoma cell line), but K3-SPG does not associate with CD3e, which is taken up by non-tumor cells. It showed that. Nanoparticles choose to be taken up by phagocytic cells such as macrophages and dendritic cells (DC), and these cells can be labeled with TRITC-dextran in vivo. Therefore, the inventor injected TRITC-dextran intravenously with fluorescently stained K3, K3-SPG, or SPG and tested for their coexistence by IHC (FIGS. 4d, e, f). i. v.
- K3-SPGs do not associate with dextran and we speculate that they accumulated passively in the space within the tumor tissue via the vascular permeability and retention (EPR) effect. ing. K3-SPG i. v.
- EPR vascular permeability and retention
- K3-SPG i. v. To test the importance of phagocytes for treatment, we injected clodronate liposomes intravenously. The present inventors usually used 100 nm clodronate liposomes instead of 200-300 nm liposomes and injected them to deplete phagocytes in the tumor (Pante, N. et al. Molecular biology). of the cell 13,425-434 (2002); Pante, N. et al.
- Cytokines such as IL-12 and IFN-I include C3-SPG (Kobiyama, K., et al. Proc. Natl. Acad. Sci. USA 111, 3086-3091 (2014)).
- ODN Korean, AM, et al. Journal of immunology 161, 428-2434 (1998); Klinman, DM, et al. Immunity 11, 123-129 (1999); Ishii, KJ. , Et al. Current opinion in molecular therapeutics 6, 166-174 (2004)). Therefore, we tested whether IL-12 and IFN-I are required for tumor reduction by treatment with K3-SPG.
- Ll7p40 and IFNAR2-deficient mice were inoculated subcutaneously with EG7 cells on day 0 and i.p. three times with PBS or K3-SPG (10 ⁇ g) as in FIG. 2 (AB). v. Treated. Thereafter, the effect of K3-SPG on tumor shrinkage was observed. (result)
- Example 4 K3-SPG treatment induces tumor cell death in a manner dependent on both IL12p40 and IFN-I
- K3-SPG treatment was demonstrated to induce tumor cell death in a manner that is dependent on both IL12p40 and IFN-I.
- Example 5 Released tumor dead cells induce anti-tumor CTL against multiple tumor antigens.
- CD8 T cells are important effectors in K3-SPG-mediated tumor shrinkage
- the results of Rag2 mice showed that the tumor suppressive effect of K3-SPG is also dependent on the adaptive immune response. Therefore, we tested CD8 T cells required for K3-SPG treatment.
- CD8 T cell depletion markedly abrogated the antitumor effect of K3-SPG (FIG. 10a (FIG. 10A)), indicating that CD8 T cells are important effector cells in this K3-SPG treatment Yes.
- Tumor reduction by K3-SPG also depends on Batf3 (deficient in cross-presented CD8 ⁇ + DC) (FIG. 10b (FIG.
- K3-SPG monotherapy is CD8 ⁇ + DC-mediated crossover. It also shows that presentation has been enhanced.
- the inventors observed a clear link between tumor invasion of CD8 T cells and tumor growth.
- CD8 T cells are expressed as K3-SPG i. v. Accumulated in the tumor area in the group i. d. There was no accumulation in the group (FIG. 10c (FIG. 10A)).
- CD8 T cells were purified from the spleens of these mice, stained with Xenlight DiR®, and K3-SPG treated (day 7, 9, and 11) another EG7 possessed The distribution of CD8 T cells labeled with Xenolight DiR® was analyzed by IVIS on day 15 (FIG. 11).
- CD8 T cells from donor mice bearing untreated tumors did not accumulate at the tumor site of WT recipient mice even when treated with K3-SPG (FIG. 10d (FIG. 10B), II).
- CD8 T cells from donor mice bearing tumors treated with K3-SPG were detected at the tumor site of recipient mice (FIG. 10d (FIG. 10B), I), and K3-SPG monotherapy was We showed that we induced anti-tumor CD8 T cells that could migrate to and invade the tumor microenvironment. These in vivo activated CD8 T cells were able to enter the tumor microenvironment of DKO recipient mice (FIG. 10e (FIG. 10B)).
- the present inventors have shown the possibility of a novel cancer immunotherapy.
- This is a new treatment where CpG is targeted to phagocytes in the tumor microenvironment (FIG. 12).
- CpG Through stimulation of TLR9, CpG induces an immune response by immune cells, and in particular activates macrophages and DCs (Klinman, DM, et al. Immunity 11, 123-129 (1999); Ishii, K J., et al. Current opinion in molecular therapeutics 6, 166-174 (2004)). This activation is very important for the anti-cancer immune response.
- phagocytes phagocytosed with this novel CpG in the tumor environment produce cytokines such as IFN and IL-12. It is very important that these cytokines are induced in the tumor environment.
- IFN ⁇ treatment directly targeting the tumor environment reactivates CTL by moving dendritic cells into the tumor and increasing antigen cross-presentation within the tumor microenvironment.
- These cytokines cause tumor cell death.
- the present inventors have found that this effect is exerted by activation of innate immunity. This cell death plays a very important role. This was a link between innate and adaptive immunity. Tumor cell death is released from the tumor microenvironment, thereby inducing acquired immunity.
- This immunogenic tumor cell death induces multiple cytotoxic T lymphocytes.
- tumor-specifically induced CTL in vivo can invade the tumor microenvironment in response to the tumor.
- This anti-tumor immune system may use endogenous antigens to address immune editing, a barrier to cancer immunotherapy.
- the circulation of tumor cells after K3-SPG single agent treatment can function as a biomarker with excellent treatment effect on tumors.
- Example 6 Formulation example
- SEQ ID NO: 2 K3-dA 40
- SPG 15 mg
- 0.25N NaOH 0.25N NaOH
- a 1 mL volume of 330 mM NaH 2 PO 4 was added to the DNA solution and then the SPG solution was added to the DNA / NaH 2 PO 4 solution and maintained at 4 ° C. overnight to complete the complexation.
- the molar ratio (M SPG / M DNA ) can be prepared by fixing at 0.27.
- Drugs used in the formulation can be obtained from Gene Design, Invivogen, Wako and others.
- the present invention provides a new form of anticancer agent that can be used as a single agent. Therefore, the complex of the present invention is useful in the pharmaceutical field as an anticancer agent.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Engineering & Computer Science (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Immunology (AREA)
- Microbiology (AREA)
- Mycology (AREA)
- Medicinal Preparation (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
Abstract
Description
(1)(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、複合体を含む抗がん剤。
(2)前記抗がん剤は、がん抗原なしで投与されることを特徴とする、項目(1)に記載の抗がん剤。
(3)前記抗がん剤は、細網内皮系および/またはリンパ節に送達されるように投与されることを特徴とする、項目(1)または(2)に記載の抗がん剤。
(4)前記細網内皮系および/またはリンパ節は、腫瘍および貪食細胞を含む、項目(3)に記載の抗がん剤。
(5)前記細網内皮系は脾臓および/または肝臓を含む、項目(3)または(4)に記載の抗がん剤。
(6)前記抗がん剤は、がん抗原なしで投与されることを特徴とする、項目(1)~(5)のいずれか1項に記載の抗がん剤。
(7)前記投与は全身性投与を含む、項目(2)~(6)のいずれか1項に記載の抗がん剤。
(8)前記全身性投与は、静脈内投与、腹腔内投与、経口投与、皮下投与、筋肉内投与、および腫瘍内投与から選択される、項目(7)に記載の抗がん剤。
(9)前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号12→7)からなる群より選択される、項目1~8のいずれか1項に記載の抗がん剤。
(10)前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、項目1~9のいずれか1項に記載の抗癌剤。
(11)前記複合体は、K3−SPGである、項目1~10のいずれか1項に記載の抗がん剤。
(細網内皮系(脾臓および/または肝臓を含む)および/またはリンパ節集積剤)
(12)(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、複合体
を含む、がんの死細胞を脾臓に集積させるための組成物。
(13)前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号7)からなる群より選択される、項目(12)に記載の組成物。
(14)前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、項目(12)または(13)に記載の組成物。
(15)前記複合体は、K3−SPGである、項目(12)~(14)のいずれか1項に記載の組成物。
(16) 前記細網内皮系および/またはリンパ節は、腫瘍および貪食細胞を含む、項目12~15のいずれか1項に記載の組成物。
(17) 前記細網内皮系は脾臓および/または肝臓を含む、項目(12)~(16)のいずれか1項に記載の組成物。
(18)前記投与は全身性投与を含む、項目(12)~(17)のいずれか1項に記載の組成物。
(19)前記全身性投与は、静脈内投与、腹腔内投与、経口投与、皮下投与、筋肉内投与、および腫瘍内投与から選択される、項目(18)に記載の組成物。
<インターロイキン12(IL12)および/またはインターフェロン(IFN)γの発現またはその促進のための組成物>
(20)(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、インターロイキン12(IL12)および/またはインターフェロン(IFN)γの発現またはその促進のための組成物。
(21)前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号7)である、項目(20)に記載の組成物。
(22)前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、項目(20)または(21)に記載の組成物。
(23)前記複合体は、K3−SPGである、項目(20)~(22)のいずれか1項に記載の組成物。 (Anticancer agent alone)
(1) (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is arranged on the 3 ′ side of the humanized K-type CpG oligodeoxynucleotide An oligodeoxynucleotide,
(B) An anticancer agent containing a complex containing β-1,3-glucan.
(2) The anticancer agent according to item (1), wherein the anticancer agent is administered without a cancer antigen.
(3) The anticancer agent according to item (1) or (2), wherein the anticancer agent is administered so as to be delivered to the reticuloendothelial system and / or lymph nodes.
(4) The anticancer agent according to item (3), wherein the reticuloendothelial system and / or lymph node includes a tumor and a phagocytic cell.
(5) The anticancer agent according to item (3) or (4), wherein the reticuloendothelial system includes spleen and / or liver.
(6) The anticancer agent according to any one of items (1) to (5), wherein the anticancer agent is administered without a cancer antigen.
(7) The anticancer agent according to any one of items (2) to (6), wherein the administration comprises systemic administration.
(8) The anticancer agent according to item (7), wherein the systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
(9) The oligodeoxynucleotides are K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5) The anticancer agent according to any one of
(10) The β-1,3-glucan is any one of
(11) The anticancer agent according to any one of
(Reticuloendothelial system (including spleen and / or liver) and / or lymph node accumulating agent)
(12) (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide An oligodeoxynucleotide,
(B) A composition for accumulating dead cells of cancer in the spleen, comprising a complex containing β-1,3-glucan.
(13) The oligodeoxynucleotide K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5) The composition according to item (12), selected from the group consisting of K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
(14) The β-1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran, according to item (12) or (13) Composition.
(15) The composition according to any one of items (12) to (14), wherein the complex is K3-SPG.
(16) The composition according to any one of
(17) The composition according to any one of items (12) to (16), wherein the reticuloendothelial system includes spleen and / or liver.
(18) The composition according to any one of items (12) to (17), wherein the administration comprises systemic administration.
(19) The composition according to item (18), wherein the systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
<Composition for expression or promotion of interleukin 12 (IL12) and / or interferon (IFN) γ>
(20) (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide An oligodeoxynucleotide,
(B) A composition for expression or promotion of interleukin 12 (IL12) and / or interferon (IFN) γ, comprising β-1,3-glucan.
(21) The oligodeoxynucleotides are K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5) The composition according to item (20), which is K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
(22) The β-1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran, according to item (20) or (21) Composition.
(23) The composition according to any one of items (20) to (22), wherein the complex is K3-SPG.
本発明のオリゴデオキシヌクレオチドに含まれるK型CpG ODNは、好ましくはヒト化されている。「ヒト化」とは、ヒトTLR9に対するアゴニスト活性を有することを意味する。従って、ヒト化K型CpG ODNを含む本発明のオリゴデオキシヌクレオチドは、ヒトに対してK型CpG ODNに特有の免疫賦活活性(例えば、ヒトB細胞を活性化してIL−6を産生させる活性)を有する。本発明において好適に用いられるK型CpG ODNは、10ヌクレオチド以上の長さであり、且つ式: K-type CpG ODNs contain multiple unmethylated CpG motifs, typically non-palindromic, and activate B cells to produce IL-6, but plasmacytoid dendritic cells (pDCs) It is a CpG ODN having structural and functional properties that hardly induce IFN-α production. An unmethylated CpG motif refers to a short nucleotide sequence containing at least one cytosine (C) -guanine (G) sequence, wherein the cytosine 5-position in the cytosine-guanine sequence is not methylated. In the following description, CpG means unmethylated CpG unless otherwise specified. Therefore, the oligodeoxynucleotide of the present invention contains K-type CpG ODN, thereby activating immunostimulatory activity peculiar to K-type CpG ODN (for example, B cells (preferably human B cells) and IL-6 Activity). A number of humanized K-type CpG ODNs are known in the art (Journal of immunology 166, 2372-2377 (2001); Journal of immunology 164, 944-953 (2000); US 8,030,285B2).
The K-type CpG ODN contained in the oligodeoxynucleotide of the present invention is preferably humanized. “Humanized” means having agonist activity against human TLR9. Therefore, the oligodeoxynucleotide of the present invention containing humanized K-type CpG ODN has an immunostimulatory activity peculiar to K-type CpG ODN (for example, an activity to activate human B cells to produce IL-6). Have The K-type CpG ODN suitably used in the present invention has a length of 10 nucleotides or more and has the formula:
本発明は、上記本発明のオリゴデオキシヌクレオチド又は上記本発明の複合体を含む、医薬組成物を提供するものである。本発明の医薬組成物は、上記本発明のオリゴデオキシヌクレオチド又は上記本発明の複合体を常套手段に従って製剤化することにより得ることができる。本発明の医薬組成物は、本発明のオリゴデオキシヌクレオチド又は複合体と薬理学的に許容され得る担体を含む。また、本医薬組成物は抗原を更に含んでいても良い。このような医薬組成物は、経口又は非経口投与に適する剤形として提供される。 (Pharmaceutical composition)
The present invention provides a pharmaceutical composition comprising the oligodeoxynucleotide of the present invention or the complex of the present invention. The pharmaceutical composition of the present invention can be obtained by formulating the oligodeoxynucleotide of the present invention or the complex of the present invention according to conventional means. The pharmaceutical composition of the present invention comprises the oligodeoxynucleotide or complex of the present invention and a pharmacologically acceptable carrier. The pharmaceutical composition may further contain an antigen. Such a pharmaceutical composition is provided as a dosage form suitable for oral or parenteral administration.
本発明のオリゴデオキシヌクレオチド及び複合体は、単独で抗がん作用を有することが見出された。このような効果は、アジュバント剤として開発されてきた本発明の特性からは予想外の効果であるといえる。それゆえ、これまでのアジュバントとしての使用のされ方、すなわち、がん抗原とともに投与することを必要とすることなく、しかも、特定のがん種に限定されることなく、汎用的な抗がん剤として身体にマイルドに作用する抗がん剤が提供されることになる。また、免疫賦活活性ももちろん有することから、他の疾患に対する免役賦活活性も期待され、体力の弱ったがん患者に対して相乗的な効果を有することも期待される。 (Medical use)
The oligodeoxynucleotides and conjugates of the present invention have been found to have anticancer activity alone. Such an effect can be said to be an unexpected effect from the characteristics of the present invention that have been developed as an adjuvant. Therefore, it can be used as an adjuvant so far, that is, it is not necessary to administer with a cancer antigen, and it is not limited to a specific cancer type. An anticancer agent that acts mildly on the body as an agent will be provided. In addition, since it also has immunostimulatory activity, it can also be expected to have immune activation activity against other diseases, and to have a synergistic effect on cancer patients with weak physical strength.
以下に本発明の好ましい実施形態を説明する。以下に提供される実施形態は、本発明のよりよい理解のために提供されるものであり、本発明の範囲は以下の記載に限定されるべきでないことが理解される。従って、当業者は、本明細書中の記載を参酌して、本発明の範囲内で適宜改変を行うことができることは明らかである。また、本発明の以下の実施形態は単独でも使用されあるいはそれらを組み合わせて使用することができることが理解される。 (Aspects of preferred embodiments)
Hereinafter, preferred embodiments of the present invention will be described. The embodiments provided below are provided for a better understanding of the present invention, and it is understood that the scope of the present invention should not be limited to the following description. Therefore, it is obvious that those skilled in the art can make appropriate modifications within the scope of the present invention with reference to the description in the present specification. It will also be appreciated that the following embodiments of the invention may be used alone or in combination.
1つの局面では、本発明は、(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、(b)β—1,3−グルカンとを含む、複合体を含む抗がん剤を提供する。本発明では、本発明の複合体自体が抗がん剤として作用することを見出した。従来は、この複合体は、アジュバントとして用いられることを本発明者らが見出し出願したにすぎず、直接、単剤として抗がん剤として用いることができるとは予想していなかった。したがって、がん抗原なしで使用されるという点で予想外の効果をもたらすといえる。 <Single agent form>
In one aspect, the invention provides an oligodeoxynucleotide comprising (a) a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide. Provided is an anticancer agent containing a complex comprising oligodeoxynucleotides arranged on the 3 ′ side and (b) β-1,3-glucan. In the present invention, it was found that the complex itself of the present invention acts as an anticancer agent. Conventionally, the present inventors have only found out and applied for the use of this complex as an adjuvant, and have not anticipated that it can be directly used as an anticancer agent as a single agent. Therefore, it can be said that it produces an unexpected effect in that it is used without a cancer antigen.
別の局面において、本発明は、(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、(b)β—1,3−グルカンとを含む、複合体を含む、がんの死細胞を細網内皮系(脾臓および/または肝臓を含む)および/またはリンパ節に集積させるための組成物を提供する。理論に束縛されることを望まないが、本発明の複合体は、がんの死細胞を細網内皮系(脾臓および/または肝臓を含む)および/またはリンパ節に集積させることができることを見出した。実施例で例証されているように、K3−SPGのような本発明の複合体による処置は、IL12p40およびIFN−Iの両方に依存する態様で、腫瘍細胞死を誘発することが実証された。このような作用を複合体が有することは従来予想されておらず、その意味で予想外の作用効果が達成されたといえる。すなわち、CpGが腫瘍微小環境における食細胞に標的化されるというものである。がんの死細胞が細網内皮系(脾臓および/または肝臓を含む)および/またはリンパ節に集積されると、その後、放出された腫瘍死細胞は複数の腫瘍抗原に対する抗腫瘍CTLを誘発し、身体にあるがん細胞が散弾銃で攻撃されるかのように殺傷され、根治させることも可能である。理論に束縛されることを望まないが、腫瘍微小環境におけるIL12およびIFN−Iサイトカインの両方を産生することは、K3−SPG単剤治療に必須とまではいえないが、重要である。 <Reticuloendothelial system (including spleen and / or liver) and / or lymph node accumulation agent>
In another aspect, the present invention provides an oligodeoxynucleotide comprising (a) a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide. 3'-side oligodeoxynucleotide and (b) β-1,3-glucan-containing complex, cancerous dead cells containing reticuloendothelial system (spleen and / or liver) And / or a composition for accumulation in lymph nodes. While not wishing to be bound by theory, it has been found that the complex of the present invention can accumulate cancer dead cells in the reticuloendothelial system (including spleen and / or liver) and / or lymph nodes. It was. As illustrated in the Examples, treatment with a complex of the invention such as K3-SPG has been demonstrated to induce tumor cell death in a manner that is dependent on both IL12p40 and IFN-I. It has not been conventionally expected that the complex has such an action, and in that sense, it can be said that an unexpected action and effect has been achieved. That is, CpG is targeted to phagocytic cells in the tumor microenvironment. Once cancer dead cells accumulate in the reticuloendothelial system (including spleen and / or liver) and / or lymph nodes, the released tumor dead cells then induce anti-tumor CTL against multiple tumor antigens. The cancer cells in the body can be killed and cured as if they were attacked with a shotgun. Without wishing to be bound by theory, it is important, although not necessarily essential for K3-SPG monotherapy, to produce both IL12 and IFN-I cytokines in the tumor microenvironment.
さらに別の局面では、本発明は、(a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、(b)β—1,3−グルカンとを含む、インターロイキン12(IL12)および/またはインターフェロン(IFN)γの発現またはその促進のための組成物を提供する。腫瘍微小環境におけるIL12およびIFN−Iサイトカインの両方を産生することは、K3−SPG単剤治療における重要な作用効果であり、このような効果は、抗がん剤としての作用のほか、他の用途においても重要である。そのような処置の対象としては、がんのほか、ウイルスなどの慢性感染症疾患、ウイルス感染予防などを挙げることができるがそれらに限定されない。 <IL12 and / or IFN expression promoter>
In yet another aspect, the present invention provides (a) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is a humanized K-type CpG oligodeoxynucleotide. For expression or promotion of interleukin 12 (IL12) and / or interferon (IFN) γ, which includes an oligodeoxynucleotide and (b) β-1,3-glucan arranged on the 3 ′ side of A composition is provided. Producing both IL12 and IFN-I cytokines in the tumor microenvironment is an important effect in K3-SPG monotherapy, such as acting as an anti-cancer agent and other It is also important in applications. Examples of such treatment include, but are not limited to, cancer, chronic infectious diseases such as viruses, and prevention of viral infection.
本発明は、上記種々の形態の医薬(治療薬または予防薬)として提供される。 (Pharmaceuticals, dosage forms, etc.)
The present invention is provided as the above-mentioned various forms of medicaments (therapeutic or preventive).
本明細書において用いられる分子生物学的手法、生化学的手法、微生物学的手法は、当該分野において周知であり慣用されるものであり、例えば、Sambrook J.et al.(1989).Molecular Cloning: A Laboratory Manual,Cold Spring Harborおよびその3rd Ed.(2001); Ausubel,F.M.(1987).Current Protocols in Molecular Biology,Greene Pub.Associates and Wiley−Interscience; Ausubel,F.M.(1989).Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology,Greene Pub.Associates and Wiley−Interscience; Innis,M.A.(1990).PCR Protocols: A Guide to Methods and Applications,Academic Press; Ausubel,F.M.(1992).Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology,Greene Pub.Associates; Ausubel,F.M. (1995).Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology,Greene Pub.Associates; Innis,M.A.et al.(1995).PCR Strategies,Academic Press;Ausubel,F.M.(1999).Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology,Wiley,and annual updates;Sninsky,J.J.et al.(1999).PCR Applications: Protocols for Functional Genomics,Academic Press、別冊実験医学「遺伝子導入&発現解析実験法」羊土社、1997などに記載されており、これらは本明細書において関連する部分(全部であり得る)が参考として援用される。 (General technology)
Molecular biological techniques, biochemical techniques, and microbiological techniques used in this specification are well known and commonly used in the art, and are described in, for example, Sambrook J. et al. et al. (1989). Molecular Cloning: A Laboratory Manual, Cold Spring Harbor and its 3rd Ed. (2001); Ausubel, F .; M.M. (1987). Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley-Interscience; Ausubel, F .; M.M. (1989). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates and Wiley-Interscience; A. (1990). PCR Protocols: A Guide to Methods and Applications, Academic Press; Ausubel, F .; M.M. (1992). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates; Ausubel, F .; M.M. (1995). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Greene Pub. Associates; Innis, M .; A. et al. (1995). PCR Strategies, Academic Press; Ausubel, F .; M.M. (1999). Short Protocols in Molecular Biology: A Compendium of Methods from Current Protocols in Molecular Biology, Wiley, and annual updates; Sninsky. J. et al. et al. (1999). PCR Applications: Protocols for Functional Genomics, Academic Press, “Experimental Methods for Gene Transfer & Expression Analysis”, Yodosha, 1997, etc., which are related in this specification (may be all) Is incorporated by reference.
以下のCpG ODNsは、(株)ジーンデザインで合成された(下線はホスホロチオエート結合を示す)。 (Production Example)
The following CpG ODNs were synthesized by Gene Design Co., Ltd. (underlined indicates phosphorothioate linkage).
7.22mgのK3−dA40を水(3.7mL)に溶解した。SPG(三井製糖)15mgを0.25N NaOH(1mL)に溶解した。1mLの330mM NaH2PO4をDNA溶液に加え、次にSPG溶液をDNA/NaH2PO4溶液に加え、4℃にて一晩維持することにより複合体化を完了した。モル比(MSPG/MDNA)は、0.27に固定した。複合体の形成は、マイクロチップ電気泳動装置(SHIMADZU:MultiNA)によって確認した複合体の形成は、サイズ排除クロマトグラフィーを用い、CpG ODNの高分子量側へのシフトを、260nmにおける吸収をモニタリングすることによって確認した(System:Agilent 1100series、Column:Asahipak GF7M−HQ(Shodex)2本連結、Flow rate:0.8mL/min、Buffer:10mM EDTA PBS,pH7.4、Temperature:40°C)。 Complexation of CpG ODN and SPG (Production Example Fig. 1)
7.22 mg of K3-dA40 was dissolved in water (3.7 mL). 15 mg of SPG (Mitsui Sugar) was dissolved in 0.25N NaOH (1 mL). Complexation was completed by adding 1 mL of 330 mM NaH2PO4 to the DNA solution and then adding the SPG solution to the DNA / NaH2PO4 solution and maintaining at 4 ° C. overnight. The molar ratio (MSPG / MDNA) was fixed at 0.27. The formation of the complex was confirmed by a microchip electrophoresis apparatus (SHIMADZU: MultiNA). The formation of the complex was performed by using size exclusion chromatography and monitoring the shift of CpG ODN toward high molecular weight at 260 nm. (System: Agilent 1100 series, Column: Asahipak GF7M-HQ (Shodex) 2 ligation, Flow rate: 0.8 mL / min, Buffer: 10 mM EDTA PBS, pH 7.4, Temperature: 40 ° C.).
以下の実施例において、強い腫瘍縮小を誘発する腫瘍微小環境における貪食細胞を標的とするナノ粒子状TLR9アゴニストによる全身性の単剤治療が可能であることを示した。 (Preparation for use in examples)
In the examples below, it was shown that systemic monotherapy with nanoparticulate TLR9 agonists targeting phagocytic cells in a tumor microenvironment that induces strong tumor shrinkage is possible.
以下に、本実施例で用いられる試薬、材料、動物、細胞およびその方法について説明する。適宜、各実施例においても補充して説明する。 (Materials and methods)
Hereinafter, reagents, materials, animals, cells and methods used in this example will be described. The description will be supplemented in each embodiment as appropriate.
6週齢の雌性C57BL/6JマウスをNihon CLEAから購入した。Il12p40欠損マウスおよびBatf3欠損マウスをJackson Laboratoryから購入した。Ifnar2欠損マウス、Myd88欠損マウスおよびDectin−1欠損マウスは以前記載した通りである(Kobiyama,K.,et al.Proc.Natl.Acad.Sci.U.S.A.111,3086−3091(2014))。全ての動物実験を、医薬基盤研究所の機関ガイドラインに従って行った。K3はGene Designにより合成された。オバルブミン(OVA)を生化学工業から購入した。 (Animals and reagents)
Six week old female C57BL / 6J mice were purchased from Nihon CLEA. Il12p40-deficient mice and Batf3-deficient mice were purchased from Jackson Laboratory. Ifnar2-deficient mice, Myd88-deficient mice, and Dectin-1-deficient mice are as previously described (Kobiyama, K., et al. Proc. Natl. Acad. Sci. USA 111, 3086-3091 (2014 )). All animal experiments were performed according to the Institute of Pharmaceutical Sciences Institute guidelines. K3 was synthesized by Gene Design. Ovalbumin (OVA) was purchased from Seikagaku.
EL4およびOVA発現EL4(EG7)は、C57BL/6Jマウスの胸腺腫細胞株であり、ATCCから購入した。B16(黒色腫)をJapanese Collection of Research Bioresoursesから購入した。B16F10(黒色腫)を理研細胞バンクから購入し、MC38(結腸がん)は、F.JAMES Primus博士により提供された。Pan02(膵臓がん)をJackson’s Laboratoryから購入した。EL4、EG7、MC38、およびPan02を、完全RPMI(10%(v/v)ウシ胎児血清(FBS)、ペニシリン、およびストレプトマイシンが追加されたRPMI 1640)で培養した。B16およびB16F10を、完全DMEM(10%(v/v)ウシ胎児血清(FBS)、ペニシリン、およびストレプトマイシンが追加されたDMEM)で培養した。 (Cell line)
EL4 and OVA expressing EL4 (EG7) is a thymoma cell line of C57BL / 6J mice and was purchased from ATCC. B16 (melanoma) was purchased from the Japan Collection of Research Bioresources. B16F10 (melanoma) was purchased from Riken Cell Bank and MC38 (colon cancer) was Provided by Dr. JAMES Primus. Pan02 (pancreatic cancer) was purchased from Jackson's Laboratory. EL4, EG7, MC38, and Pan02 were cultured in complete RPMI (RPMI 1640 supplemented with 10% (v / v) fetal bovine serum (FBS), penicillin, and streptomycin). B16 and B16F10 were cultured in complete DMEM (DMEM supplemented with 10% (v / v) fetal bovine serum (FBS), penicillin, and streptomycin).
EG7、EL4、B16、B16F10、およびMC38細胞(10%マトリゲル/PBS中に5×106細胞/mLで100μl)を、マウスの右側腹部の皮下(s.c)に接種した。腫瘍サイズを、腫瘍の長さ(L)、幅(W)、および高さ(H)で測定し、腫瘍体積(V)をV=L×W×Hとして計算した。腫瘍内注射(i.t.)では、腫瘍部位に直接注射した。CpG治療は、腫瘍体積が約100mm3に達してから開始し、その時期は、EG7およびB16F10の接種の7日後、B16の接種の10日後、ならびにMC38の接種の14日後であった。腫瘍保有マウスを、K3(30μg)またはK3−SPG(10μg)で1日おきに3回処置した。 (Tumor experiment and treatment method)
EG7, EL4, B16, B16F10, and MC38 cells (100 μl at 5 × 10 6 cells / mL in 10% Matrigel / PBS) were inoculated subcutaneously (sc) in the right flank of mice. Tumor size was measured by tumor length (L), width (W), and height (H), and tumor volume (V) was calculated as V = L × W × H. Intratumoral injection (it) was injected directly into the tumor site. CpG treatment began when the tumor volume reached approximately 100 mm 3 , which was 7 days after EG7 and B16F10 inoculation, 10 days after B16 inoculation, and 14 days after MC38 inoculation. Tumor-bearing mice were treated 3 times every other day with K3 (30 μg) or K3-SPG (10 μg).
Pan02の腹膜播種モデルにおいて、1×106個のPan02細胞(PBS中に1×107細胞/mLで100μl)を、腹腔内に注射した。CpG治療を接種11日後に開始し、全ての腫瘍小結節を21日目にマウスの腹膜から摘出し、その後これらの重量(g)を測定した。CpG治療での投与量は上記のとおりである。 (Pan02 peritoneal seeding model)
In the Pan02 peritoneal seeding model, 1 × 10 6 Pan02 cells (100 μl at 1 × 10 7 cells / mL in PBS) were injected intraperitoneally. CpG treatment was started 11 days after inoculation and all tumor nodules were excised from the peritoneum of mice on day 21 and their weight (g) was then measured. The dosage for CpG treatment is as described above.
K3およびK3−SPGの局在を評価するために、C57BL/6マウスに、EG7を0日目にs.c.接種し、PBS(コントロール)、Alexa 647−K3(30μg)、またはAlexa 647−K3−SPG(10μg)を12日目にi.v.投与した。投与1時間後、マウスをIVIS(登録商標)Lumina Imaging Systemおよび分析ソフトウェア(Ver.2.6、Xenogen)で分析し、相対的蛍光で測定されたイメージを、表面放射輝度の物理単位(光子/秒/cm2/sr)に変換した。in vivoで標識されたCD8+T細胞を検出するために、脾細胞を、7、9、11日目にK3−SPGで処置されたか、または処置されていない、EG7を保有するC57BL/6マウスまたはIl12p40−Ifnar2ダブルノックアウトマウスから14日目に回収した。脾細胞の懸濁後、赤血球をACK溶解緩衝液(150mM NH4Cl、10mM KHCO3、0.1mM Na2EDTA)で溶解し、細胞を完全RPMI中に維持した。CD8α+T細胞をMACS(Miltenyi Biotec)によりソートした。CD8α+T細胞を陰性選択の方法でソートした。その後、ソートされたCD8α+T細胞を、Xenolight DiR(登録商標)で染色した。染色されたCD8α+T細胞を、14日目にレシピエントマウス(0日目にEG7を接種し、7、9、11日目にK3−SPGでi.v.処置されたか、または処置されていないC57BL/6マウスまたはIl12p40−Ifnar2ダブルノックアウトマウス)に移した。染色した細胞を移して24時間後に、マウスをIVIS(登録商標)Lumina Imaging System(Ver.2.6)で分析した。目的の領域を腫瘍領域に集約し、蛍光強度をLiving Image Software(Ver.2.6、Xenogen)で分析した。 (In vivo imaging experiment)
To assess the localization of K3 and K3-SPG, C57BL / 6 mice were treated with EG7 at
C57BL/6Jマウス(6−8週齢、雌、日本クレア)に、Alexa 647−K3(30μg)、Alexa 647−K3−SPG(10μg)、およびデキストラン−PE(20μg)を尾静脈からi.v.注射した。腫瘍を注射後1時間後に回収し、凍結切片を、4%(w/v)パラホルムアルデヒドで10分間固定し、Hoechst 33258と一緒に抗CD3e抗体、抗CD8β抗体で染色した。細胞をOlympus IX81システムを使用して撮影した。イメージデータをMetaMorphで分析した。 (Immunohistochemistry)
C57BL / 6J mice (6-8 weeks old, female, CLEA Japan) were treated with Alexa 647-K3 (30 μg), Alexa 647-K3-SPG (10 μg), and dextran-PE (20 μg) i.e. from the tail vein. v. Injected. Tumors were collected 1 hour after injection and frozen sections were fixed with 4% (w / v) paraformaldehyde for 10 minutes and stained with anti-CD3e and anti-CD8β antibodies together with
食細胞(樹状細胞およびマクロファージ)を枯渇させるために、C57BL/6マウスに、クロドロネートリポソームまたはコントロールリポソーム(100nm)(片山化学)をEG7の接種の5日後にi.v.注射した。CD8+T細胞を枯渇させるために、200μgの抗CD8α抗体を、EG7接種の6日後および13日後に尾静脈にi.v.注射した。 (Depletion experiment)
In order to deplete phagocytic cells (dendritic cells and macrophages), C57BL / 6 mice were treated with clodronate liposomes or control liposomes (100 nm) (Katayama Chemical) i. v. Injected. To deplete CD8 + T cells, 200 μg of anti-CD8α antibody was administered i.v. to the
脾細胞を、7、9、11日目にK3−SPGでi.v.処置されたか、処置されていない、EG7を保有するC57BL/6マウスまたはIl12p40−Ifnar2ダブルノックアウトマウスから14日目に回収した。脾細胞の調製後、赤血球をACK溶解緩衝液で溶解し、細胞を完全RPMIで維持した。脾細胞をH−2Kb OVAテトラマー(MBL)、抗CD8α抗体(KT15)、抗TCRβ抗体(H57−597)、抗CD62L抗体(MEL−14)、および抗CD44抗体(IM7)、ならびに7−アミノアクチノマイシンD(7AAD)で染色した。OVAテトラマー+CD44+CD8α+TCRβ+の細胞数を、フローサイトメトリーで決定した。他の実験については、調製された脾細胞を、抗CD45抗体、抗CD3e抗体、抗CD8α抗体、および抗CD11a抗体と共にインキュベートし、その後フローサイトメトリーで分析した。 (Analysis of splenocytes)
Splenocytes were injected i.p. with K3-SPG on
脾細胞を、7、9、11日目にK3−SPGでi.v.処置されたか、処置されていない、EG7を保有するC57BL/6マウスまたはIl12p40−Ifnar2ダブルノックアウトマウスから12日目に回収した。脾細胞の調製後、赤血球をACK溶解緩衝液で溶解し、細胞を完全RPMI中に維持した。脾細胞を抗CD45抗体(APC)で染色し、CD45−細胞の数をフローサイトメトリーで決定した。さらに、アポトーシス細胞、ネクローシス細胞、およびCD45陰性生細胞の集団を、PIおよびHoechst 33342で染色し、その後フローサイトメトリーで分析した。次に、CD45−細胞を、K3−SPGで処置された腫瘍保有C57BL/6マウスから、INFLUX(BD Bioscience)によってソートした。 (Assay and immunization of CD45 negative cells)
Splenocytes were injected i.p. with K3-SPG on
C57BL/6マウスに、5×105個のCD45−細胞を−7日目にi.v.投与した。免疫化7日後、マウスに5×105個のEG7細胞を0日目にs.c.接種した。 (Vaccination model)
C57BL / 6 mice were challenged with 5 × 10 5 CD45 − cells on day −7. v. Administered. Seven days after immunization, mice were challenged with 5 × 10 5 EG7 cells on
マウスIL−12p40、マウスIL−13、およびヒトIFNγのレベルを、R&DのELISAキットを用いて測定した。 (Measurement of cytokines)
Mouse IL-12p40, mouse IL-13, and human IFNγ levels were measured using an R & D ELISA kit.
Mann−WhitneyのU検定、Studentのt検定またはBonferroniの多重比較検定を含む一元分散分析を、統計分析に用いた(*p<0.05;**p<0.01;***p<0.001)。統計分析をGraphPad Prism software(La Jolla、CA、USA)を用いて行った。 (Statistical analysis)
One-way analysis of variance including Mann-Whitney U test, Student's t test or Bonferroni's multiple comparison test was used for statistical analysis ( * p <0.05; ** p <0.01; *** p < 0.001). Statistical analysis was performed using GraphPad Prism software (La Jolla, CA, USA).
本実施例では、K3−SPGの静脈内注射により、腫瘍抗原を全く追加しなくても強い腫瘍成長の抑制が誘発されることを実証した。 (Example 1: Intravenous injection of K3-SPG induces strong tumor growth inhibition without any additional tumor antigen)
In this example, it was demonstrated that intravenous injection of K3-SPG induces strong tumor growth inhibition without any additional tumor antigen.
C57BL/6マウスに、EG7(OVA発現マウス胸腺腫細胞株)を右側腹部に0日目に接種し、尾基部付近の皮下(i.d.)投与、腫瘍内(i.t.)投与、または静脈内(i.v.)投与の3つの異なる経路を介して、PBS、および等モル量のK3(30μg)またはK3−SPG(10μg)で3回処置した(接種の7、9、11日後)。腫瘍サイズを23日目まで2~3日毎に測定した。 (Experiment with EG7 (OVA expressing mouse thymoma cell line) model)
C57BL / 6 mice were inoculated with EG7 (OVA expressing mouse thymoma cell line) on the right flank on
結果は図2(A~B)以下に表す。PBSの群(コントロール)において、腫瘍成長はいずれの投与経路を介しても抑制されなかった(図2a、b、c(図2A))。K3処置において、腫瘍縮小がi.t.でのみ観察されたが、他の経路においては観察されなかった(図2d、e、f(図2A))。K3−SPG処置において、i.t.およびi.v.の両方で強い腫瘍縮小が観察されたが、i.d.投与は、腫瘍成長に対する影響を示さなかった(図2g、h、i(図2A))。コントロール、K3およびK3−SPGを比較して示した図を図(図2a、d、g(図2A))に示す。 (result)
The results are shown in FIG. 2 (AB) and below. In the PBS group (control), tumor growth was not inhibited via any route of administration (FIGS. 2a, b, c (FIG. 2A)). In K3 treatment, tumor shrinkage is i. t. Was observed only in, but not in other routes (FIGS. 2d, e, f (FIG. 2A)). In K3-SPG treatment, i. t. And i. v. Strong tumor shrinkage was observed in both cases, i. d. Administration showed no effect on tumor growth (Figure 2g, h, i (Figure 2A)). The figure which compared and showed control, K3, and K3-SPG is shown in a figure (FIG. 2 a, d, g (FIG. 2A)).
このK3−SPG全身性単剤治療の潜在性を調査するために、他の腫瘍細胞株もまた、EG7モデルにおいて使用された同様のプロトコルで試験した。 (Experiment with other tumor cell lines)
To investigate the potential of this K3-SPG systemic monotherapy, other tumor cell lines were also tested with similar protocols used in the EG7 model.
K3−SPGの静脈内投与は、黒色腫(B16およびB16F10)および結腸がん(MC38)の成長も抑制した(図2j、k、l(図2B))。本発明者らは、さらにより臨床的悪性度の高い腫瘍播種性モデルを作製することによって試験した。マウス膵臓腫瘍株、Pan02(1×106細胞)を、腹腔内(「i.p.」とも称する)に接種し、その後、K3またはK3−SPGによる治療(1日おきに3回)を接種の11日後に開始した。21日目に全てのマウスを屠殺し、腹腔の腫瘍の総重量を評価した(図2m(図2B))。腫瘍成長は、K3−SPG i.v.処置群において顕著に抑制されたが、K3 i.p.およびK3−SPG i.p.処置群においては抑制されなかった(図2m(図2B))。それに応じて、顕著な生存の延長が、K3−SPG i.v.処置群において観察されたが、K3 i.v.群では観察されなかった(図2n(図2B))。これらの結果は、K3−SPGの全身性i.v.投与が、多くの異なるがんに対する有望な単剤治療であり、いずれの腫瘍ペプチドおよび抗原もさらに必要としないことを実証するものである。 (result)
Intravenous administration of K3-SPG also suppressed the growth of melanoma (B16 and B16F10) and colon cancer (MC38) (Figure 2j, k, l (Figure 2B)). We tested by creating a tumor dissemination model with even higher clinical malignancy. Mouse pancreatic tumor line, Pan02 (1 × 10 6 cells) is inoculated intraperitoneally (also referred to as “ip”) followed by K3 or K3-SPG treatment (3 times every other day) 11 days later. On day 21, all mice were sacrificed and the total weight of the abdominal tumor was assessed (FIG. 2m (FIG. 2B)). Tumor growth is determined by K3-SPG i. v. Although significantly suppressed in the treatment group, K3 i. p. And K3-SPG i. p. It was not suppressed in the treatment group (FIG. 2m (FIG. 2B)). In response, a significant prolongation of survival was observed in K3-SPG i. v. Although observed in the treatment group, K3 i. v. Not observed in the group (FIG. 2n (FIG. 2B)). These results indicate that the systemic i. v. It demonstrates that administration is a promising monotherapy for many different cancers and does not require any further tumor peptides and antigens.
次に、本発明者らは、K3−SPGの腫瘍微小環境におけるメカニズムを解明した。 (Example 2: K3-SPG targeted phagocytic cells in tumor microenvironment)
Next, the present inventors elucidated the mechanism of K3-SPG in the tumor microenvironment.
in vivoでの分布を試験するために、K3およびK3−SPGを蛍光標識した。EG7腫瘍保有マウスにPBS、Alexa647−K3(30μg)、またはAlexa647−K3−SPG(10μg)をi.v.注射し、その後、蛍光の分布をin vivoイメージングシステム(IVIS)によって試験した。 (Fluorescent labeling imaging)
To test the distribution in vivo, K3 and K3-SPG were fluorescently labeled. EG7 tumor-bearing mice were given PBS, Alexa647-K3 (30 μg), or Alexa647-K3-SPG (10 μg) i. v. After injection, the distribution of fluorescence was examined by an in vivo imaging system (IVIS).
次に、本実施例では、本発明者らは、K3−SPG単剤治療の成功に必要とされる因子について試験した。 (Example 3: Producing both IL12 and IFN-I cytokines in the tumor microenvironment is important for K3-SPG monotherapy)
Next, in this example, the inventors tested for factors required for successful K3-SPG monotherapy.
(結果) Ll7p40 and IFNAR2-deficient mice were inoculated subcutaneously with EG7 cells on
(result)
本実施例では、K3−SPG処置は、IL12p40およびIFN−Iの両方に依存する態様で、腫瘍細胞死を誘発することを実証した。 Example 4: K3-SPG treatment induces tumor cell death in a manner dependent on both IL12p40 and IFN-I
In this example, K3-SPG treatment was demonstrated to induce tumor cell death in a manner that is dependent on both IL12p40 and IFN-I.
本実施例では、放出された腫瘍死細胞は複数の腫瘍抗原に対する抗腫瘍CTLを誘発することを実証した。 (Example 5: Released tumor dead cells induce anti-tumor CTL against multiple tumor antigens)
In this example, it was demonstrated that released tumor dead cells induce anti-tumor CTL against multiple tumor antigens.
Rag2マウスの結果は、K3−SPGの腫瘍抑制効果もまた、適応免疫応答に依存することを示した。それゆえ、本発明者らは、K3−SPG治療に必要なCD8T細胞を試験した。in vivoにおけるCD8 T細胞の枯渇は、K3−SPGの抗腫瘍効果を顕著に抑止し(図10a(図10A))、CD8T細胞がこのK3−SPG治療における重要なエフェクター細胞であることを示している。また、K3−SPGによる腫瘍縮小は、Batf3(交差提示CD8α+ DCを欠損している)にも依存し(図10b(図10A))、K3−SPG単剤治療が、CD8α+ DC媒介性交差提示をも増強したことを示している。本発明者らは、CD8T細胞の腫瘍浸潤と腫瘍成長との間の明らかな関連性を観察した。CD8T細胞は、K3−SPG i.v.群において腫瘍領域で蓄積したが、i.d.群では蓄積しなかった(図10c(図10A))。 (CD8 T cells are important effectors in K3-SPG-mediated tumor shrinkage)
The results of Rag2 mice showed that the tumor suppressive effect of K3-SPG is also dependent on the adaptive immune response. Therefore, we tested CD8 T cells required for K3-SPG treatment. In vivo CD8 T cell depletion markedly abrogated the antitumor effect of K3-SPG (FIG. 10a (FIG. 10A)), indicating that CD8 T cells are important effector cells in this K3-SPG treatment Yes. Tumor reduction by K3-SPG also depends on Batf3 (deficient in cross-presented CD8α + DC) (FIG. 10b (FIG. 10A)), and K3-SPG monotherapy is CD8α + DC-mediated crossover. It also shows that presentation has been enhanced. The inventors observed a clear link between tumor invasion of CD8 T cells and tumor growth. CD8 T cells are expressed as K3-SPG i. v. Accumulated in the tumor area in the group i. d. There was no accumulation in the group (FIG. 10c (FIG. 10A)).
(考察) Finally, we tested what is necessary for these CD8 T cells to enter the tumor area. WT and Il12p40-Ifnar2 DKO mice are inoculated with EG7 cells on
(Discussion)
製剤化する場合の組成をご教示ください
製剤は、例えば、7.22mgのK3−dA40(配列番号2)を水(3.7mL)に溶解し、SPG(15mg)を0.25N NaOH(1mL)に溶解した。1mLの容積の330mM NaH2PO4をDNA溶液に加え、次いで、SPG溶液をこのDNA/NaH2PO4溶液に加え、4℃で一晩維持して、複合体化を完了させた。モル比(MSPG/MDNA)は0.27に固定して製造することができる。 (Example 6: Formulation example)
Please tell me the composition when formulating. For example, 7.22 mg of K3-dA 40 (SEQ ID NO: 2) is dissolved in water (3.7 mL), and SPG (15 mg) is dissolved in 0.25N NaOH (1 mL). ). A 1 mL volume of 330 mM NaH 2 PO 4 was added to the DNA solution and then the SPG solution was added to the DNA / NaH 2 PO 4 solution and maintained at 4 ° C. overnight to complete the complexation. The molar ratio (M SPG / M DNA ) can be prepared by fixing at 0.27.
配列番号2:K3−dA40
配列番号3:dA40−K3
配列番号4:K3−dA20
配列番号5:K3−dA25
配列番号6:K3−dA30
配列番号7:K3−dA35
SEQ ID NO: 1 K3
SEQ ID NO: 2: K3-dA 40
SEQ ID NO: 3: dA 40 -K3
SEQ ID NO: 4: K3-dA20
SEQ ID NO: 5: K3-dA25
SEQ ID NO: 6: K3-dA30
SEQ ID NO: 7: K3-dA35
Claims (23)
- (a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、複合体を含む抗がん剤。 (A) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide; Oligodeoxynucleotides;
(B) An anticancer agent containing a complex containing β-1,3-glucan. - 前記抗がん剤は、がん抗原なしで投与されることを特徴とする、請求項1に記載の抗がん剤。 The anticancer agent according to claim 1, wherein the anticancer agent is administered without a cancer antigen.
- 前記抗がん剤は、細網内皮系および/またはリンパ節に送達されるように投与されることを特徴とする、請求項1または2に記載の抗がん剤。 The anticancer agent according to claim 1 or 2, wherein the anticancer agent is administered so as to be delivered to a reticuloendothelial system and / or a lymph node.
- 前記細網内皮系および/またはリンパ節は、腫瘍および貪食細胞を含む、請求項3に記載の抗がん剤。 The anticancer agent according to claim 3, wherein the reticuloendothelial system and / or lymph node includes tumor and phagocytic cells.
- 前記細網内皮系は脾臓および/または肝臓を含む、請求項3または4に記載の抗がん剤。 The anticancer agent according to claim 3 or 4, wherein the reticuloendothelial system includes spleen and / or liver.
- 前記抗がん剤は、がん抗原なしで投与されることを特徴とする、請求項1~5のいずれか1項に記載の抗がん剤。 The anticancer agent according to any one of claims 1 to 5, wherein the anticancer agent is administered without a cancer antigen.
- 前記投与は全身性投与を含む、請求項2~6のいずれか1項に記載の抗がん剤。 The anticancer agent according to any one of claims 2 to 6, wherein the administration comprises systemic administration.
- 前記全身性投与は、静脈内投与、腹腔内投与、経口投与、皮下投与、筋肉内投与、および腫瘍内投与から選択される、請求項7に記載の抗がん剤。 The anticancer agent according to claim 7, wherein the systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
- 前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号7)からなる群より選択される、請求項1~8のいずれか1項に記載の抗がん剤。 The oligodeoxynucleotide is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5), The anticancer agent according to any one of claims 1 to 8, which is selected from the group consisting of K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- 前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、請求項1~9のいずれか1項に記載の抗癌剤。 The anticancer agent according to any one of claims 1 to 9, wherein the β-1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran, and laminaran. .
- 前記複合体は、K3−SPGである、請求項1~10のいずれか1項に記載の抗がん剤。 The anticancer agent according to any one of claims 1 to 10, wherein the complex is K3-SPG.
- (a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、複合体
を含む、がんの死細胞を細網内皮系および/またはリンパ節に集積させるための組成物。 (A) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide; Oligodeoxynucleotides;
(B) A composition for accumulating dead cells of cancer in the reticuloendothelial system and / or lymph nodes, comprising a complex containing β-1,3-glucan. - 前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号7)からなる群より選択される、請求項12に記載の組成物。 The oligodeoxynucleotide is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5), 13. The composition of claim 12, wherein the composition is selected from the group consisting of K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- 前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、請求項12または13に記載の組成物。 The composition according to claim 12 or 13, wherein the β-1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran.
- 前記複合体は、K3−SPGである、請求項12~14のいずれか1項に記載の組成物。 The composition according to any one of claims 12 to 14, wherein the complex is K3-SPG.
- 前記細網内皮系および/またはリンパ節は、腫瘍および貪食細胞を含む、請求項12~15のいずれか1項に記載の組成物。 The composition according to any one of claims 12 to 15, wherein the reticuloendothelial system and / or lymph node contains a tumor and a phagocytic cell.
- 前記細網内皮系は脾臓および/または肝臓を含む、請求項12~16のいずれか1項に記載の組成物。 The composition according to any one of claims 12 to 16, wherein the reticuloendothelial system includes spleen and / or liver.
- 前記投与は全身性投与を含む、請求項12~17のいずれか1項に記載の組成物。 The composition according to any one of claims 12 to 17, wherein the administration comprises systemic administration.
- 前記全身性投与は、静脈内投与、腹腔内投与、経口投与、皮下投与、筋肉内投与、および腫瘍内投与から選択される、請求項18に記載の組成物。 19. The composition of claim 18, wherein the systemic administration is selected from intravenous administration, intraperitoneal administration, oral administration, subcutaneous administration, intramuscular administration, and intratumoral administration.
- (a)ヒト化K型CpGオリゴデオキシヌクレオチド及びポリデオキシアデニル酸を含む、オリゴデオキシヌクレオチドであって、ポリデオキシアデニル酸が、ヒト化K型CpGオリゴデオキシヌクレオチドの3’側に配置されている、オリゴデオキシヌクレオチドと、
(b)β—1,3−グルカンとを
含む、インターロイキン12(IL12)および/またはインターフェロン(IFN)γの発現またはその促進のための組成物。 (A) an oligodeoxynucleotide comprising a humanized K-type CpG oligodeoxynucleotide and polydeoxyadenylic acid, wherein the polydeoxyadenylic acid is located 3 ′ of the humanized K-type CpG oligodeoxynucleotide; Oligodeoxynucleotides;
(B) A composition for expression or promotion of interleukin 12 (IL12) and / or interferon (IFN) γ, comprising β-1,3-glucan. - 前記オリゴデオキシヌクレオチドはK3(配列番号1)、K3−dA40(配列番号2)、dA40−K3(配列番号3)、K3−dA20(配列番号4)、K3−dA25(配列番号5)、K3−dA30(配列番号6)およびK3−dA35(配列番号7)である、請求項20に記載の組成物。 The oligodeoxynucleotide is K3 (SEQ ID NO: 1), K3-dA 40 (SEQ ID NO: 2), dA 40 -K3 (SEQ ID NO: 3), K3-dA20 (SEQ ID NO: 4), K3-dA25 (SEQ ID NO: 5), 21. The composition of claim 20, which is K3-dA30 (SEQ ID NO: 6) and K3-dA35 (SEQ ID NO: 7).
- 前記β—1,3−グルカンはシゾフィラン(SPG)、レンチナン、スクレログルカン、カードラン、パーキマン、グリホランおよびラミナランからなる群より選択される、請求項20または21に記載の組成物。 The composition according to claim 20 or 21, wherein the β-1,3-glucan is selected from the group consisting of schizophyllan (SPG), lentinan, scleroglucan, curdlan, parkan, glyphoran and laminaran.
- 前記複合体は、K3−SPGである、請求項20~22のいずれか1項に記載の組成物。 The composition according to any one of claims 20 to 22, wherein the complex is K3-SPG.
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US15/539,627 US20180256630A1 (en) | 2014-12-26 | 2014-12-26 | Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug |
PCT/JP2014/084772 WO2016103531A1 (en) | 2014-12-26 | 2014-12-26 | Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug |
JP2016565861A JP6536964B2 (en) | 2014-12-26 | 2014-12-26 | Application of nucleic acid polysaccharide complex having immunostimulatory activity as antitumor agent |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
PCT/JP2014/084772 WO2016103531A1 (en) | 2014-12-26 | 2014-12-26 | Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2016103531A1 true WO2016103531A1 (en) | 2016-06-30 |
Family
ID=56149611
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2014/084772 WO2016103531A1 (en) | 2014-12-26 | 2014-12-26 | Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug |
Country Status (3)
Country | Link |
---|---|
US (1) | US20180256630A1 (en) |
JP (1) | JP6536964B2 (en) |
WO (1) | WO2016103531A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018174140A1 (en) | 2017-03-23 | 2018-09-27 | ナパジェン ファーマ,インコーポレテッド | Cancer cell adhesion activity inhibitor |
WO2021132528A1 (en) * | 2019-12-25 | 2021-07-01 | ナパジェン ファーマ,インコーポレテッド | Short-chain cpg-containing oligodeoxynucleotide with linked polydeoxyadenylic acid, complex containing said oligodeoxynucleotide, and use thereof |
WO2022102694A1 (en) | 2020-11-12 | 2022-05-19 | 第一三共株式会社 | COMPLEX OF β-GLUCAN AND NUCLEIC ACID HAVING CONTROLLED PARTICLE SIZE |
Families Citing this family (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN113041256A (en) * | 2019-12-26 | 2021-06-29 | 南京绿叶制药有限公司 | Combined drug delivery system, method and application using CpG oligodeoxynucleotide and beta-1, 3-glucan |
Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2008100919A (en) * | 2006-10-17 | 2008-05-01 | Japan Science & Technology Agency | Nucleic acid/polysaccharide complex useful for preventing th2 cell-associated disease |
Family Cites Families (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US6207646B1 (en) * | 1994-07-15 | 2001-03-27 | University Of Iowa Research Foundation | Immunostimulatory nucleic acid molecules |
US8323644B2 (en) * | 2006-01-17 | 2012-12-04 | Sloan-Kettering Institute For Cancer Research | Therapy-enhancing glucan |
TW200819787A (en) * | 2006-10-27 | 2008-05-01 | Benq Corp | Light-separation apparatus |
WO2015041318A1 (en) * | 2013-09-20 | 2015-03-26 | 独立行政法人医薬基盤研究所 | Complex containing oligonucleotide having immunopotentiating activity and use thereof |
-
2014
- 2014-12-26 JP JP2016565861A patent/JP6536964B2/en active Active
- 2014-12-26 US US15/539,627 patent/US20180256630A1/en not_active Abandoned
- 2014-12-26 WO PCT/JP2014/084772 patent/WO2016103531A1/en active Application Filing
Patent Citations (1)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
JP2008100919A (en) * | 2006-10-17 | 2008-05-01 | Japan Science & Technology Agency | Nucleic acid/polysaccharide complex useful for preventing th2 cell-associated disease |
Non-Patent Citations (7)
Title |
---|
ASANO, K. ET AL.: "CD 169-Positive Macrophages Dominate Antitumor Immunity by Crosspresenting Dead Cell -Associated Antigens", IMMUNITY, vol. 34, no. 28, 2012, pages 85 - 95 * |
HUANG, Z. ET AL.: "Targeted delivery of oligonucleotides into tumor-associated macrophages for cancer immunotherapy", J. CONTROL. RELEASE, vol. 158, no. 2, 2012, pages 286 - 292 * |
KOBIYAMA, K. ET AL.: "K3-SPG is a novel nano- particulate CpG oligodeoxynucleotide complex with robust interferon induction and adjuvanticity", JAPANESE SOCIETY FOR IMMUNOLOGY SOKAI GAKUJUTSU SHUKAI KIROKU, vol. 42, 2013, pages 163 * |
SHIMADA, N. ET AL.: "A polysaccharide carrier to effectively deliver native phosphodiester CpG DNA to antigen-presenting cells", BIOCONJUG. CHEM., vol. 18, no. 4, 2007, pages 1280 - 1286, XP055145914, doi:10.1021/bc0700178 * |
YUJI KITAHATA ET AL.: "Jisedai TLR9 Agonist (K3-SPG) no Ko Shuyo Koka to sono Mechanism no Kaiseki", DAI 69 KAI THE JAPANESE SOCIETY OF GASTROENTEROLOGICAL SURGERY SOKAI SHOROKU, 7 July 2014 (2014-07-07), Retrieved from the Internet <URL:http://www.myschedule.jp/69jsgs/detail.php?sessionuniqueid=WS-008&sessid=258&stronq=1> [retrieved on 20150212] * |
YUJI KITAHATA ET AL.: "Mechanism study of nanoparticulate CpG-ODN(K3-SPG) for anti-tumor study", DAI 73 KAI ANNUAL MEETING OF THE JAPAN CANCER ASSOCIATION SHOROKU, 2014 NEN 9 GATSU, J-3047 * |
YUJI KITAHATA ET AL.: "Shuyo Kankyo o Hyoteki to shita TLR9 Agonist Toyo ni yoru Atarashii Gan Men'eki Ryoho no Kaihatsu", DAI 18 KAI JAPANESE ASSOCIATION OF CANCER IMMUNOLOGY SOKAI PROGRAM/SHOROKUSHU, 30 June 2014 (2014-06-30), pages 156 * |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2018174140A1 (en) | 2017-03-23 | 2018-09-27 | ナパジェン ファーマ,インコーポレテッド | Cancer cell adhesion activity inhibitor |
EP3610880A4 (en) * | 2017-03-23 | 2021-01-06 | Napajen Pharma, Inc. | Cancer cell adhesion activity inhibitor |
WO2021132528A1 (en) * | 2019-12-25 | 2021-07-01 | ナパジェン ファーマ,インコーポレテッド | Short-chain cpg-containing oligodeoxynucleotide with linked polydeoxyadenylic acid, complex containing said oligodeoxynucleotide, and use thereof |
WO2022102694A1 (en) | 2020-11-12 | 2022-05-19 | 第一三共株式会社 | COMPLEX OF β-GLUCAN AND NUCLEIC ACID HAVING CONTROLLED PARTICLE SIZE |
KR20230107833A (en) | 2020-11-12 | 2023-07-18 | 다이이찌 산쿄 가부시키가이샤 | Complex of beta glucan and nucleic acid with controlled particle diameter |
Also Published As
Publication number | Publication date |
---|---|
US20180256630A1 (en) | 2018-09-13 |
JPWO2016103531A1 (en) | 2017-10-05 |
JP6536964B2 (en) | 2019-07-03 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7278551B2 (en) | Oligonucleotide-containing complex having immunostimulatory activity and use thereof | |
US11242533B2 (en) | RNA-nanostructured double robots and methods of use thereof | |
JP5871212B2 (en) | Novel nucleic acid having adjuvant activity and use thereof | |
US11110168B2 (en) | Nanoparticles, controlled-release dosage forms, and methods for delivering an immunotherapeutic agent | |
WO2016103531A1 (en) | Use of nucleic acid-polysaccharide complexes having immunopotentiating activity as anti-tumor drug | |
JP6698069B2 (en) | CpG spacer oligonucleotide-containing complex having immunostimulatory activity and use thereof | |
JP6715775B2 (en) | Non-aggregating immunostimulatory oligonucleotide | |
JP7048102B2 (en) | Application of nucleic acid polysaccharide complex with immunostimulatory activity as an antitumor drug | |
TWI725494B (en) | Application of nucleic acid polysaccharide complex with immune activating activity as an antitumor drug | |
TWI701047B (en) | Application of nucleic acid polysaccharide complex with immune activating activity as an antitumor drug | |
US20230053708A1 (en) | Short-chain cpg-containing oligodeoxynucleotide with linked polydeoxyadenylic acid, complex containing said oligodeoxynucleotide, and use thereof |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 14909131 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2016565861 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 15539627 Country of ref document: US |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 14909131 Country of ref document: EP Kind code of ref document: A1 |